Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MODULATION OF ENDOPLASMIC RETICULUM AMINOPEPTIDASE 2 (ERAP2)-MEDIATED IMMUNE RESPONSE
Document Type and Number:
WIPO Patent Application WO/2022/026336
Kind Code:
A1
Abstract:
The present disclosure provides methods of treating subjects having an immune disorder by administering a therapeutically effective amount of an Endoplasmic Reticulum Aminopeptidase 2 (ERAP2) inhibitor to the subject, and optionally an Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) agonist or inhibitor and/or an HLA-Aw19 inhibitor, and also provides methods of identifying subjects having an increased risk for developing an MHC-I-opathy.

Inventors:
GELFMAN SAHAR (US)
LIGOCKI ANN (US)
COPPOLA GIOVANNI (US)
BARIS ARIS (US)
MOSCATI ARDEN (US)
STAHL ELI A (US)
KOSMICKI JACK A (US)
FERREIRA MANUEL ALLEN REVEZ (US)
ROMANO CARMELO (US)
Application Number:
PCT/US2021/043062
Publication Date:
February 03, 2022
Filing Date:
July 23, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
REGENERON PHARMA (US)
International Classes:
C07K16/40; A61K31/7088; A61P17/00; A61P19/00; A61P27/00; A61P37/02; C12N9/48
Other References:
GEORGIADIS DIMITRIS ET AL: "Inhibitors of ER Aminopeptidase 1 and 2: From Design to Clinical Application", CURRENT MEDICINAL CHEMISTRY, vol. 26, no. 15, 25 July 2019 (2019-07-25), NL, pages 2715 - 2729, XP055794307, ISSN: 0929-8673, DOI: 10.2174/0929867325666180214111849
LÓPEZ DE CASTRO JOSÉ A ET AL: "Molecular and pathogenic effects of endoplasmic reticulum aminopeptidases ERAP1 and ERAP2 in MHC-I-associated inflammatory disorders: Towards a unifying view", MOLECULAR IMMUNOLOGY, PERGAMON, GB, vol. 77, 11 August 2016 (2016-08-11), pages 193 - 204, XP029708117, ISSN: 0161-5890, DOI: 10.1016/J.MOLIMM.2016.08.005
AGRAWAL N ET AL: "Genetic associations and functional characterization of M1 aminopeptidases and immune-mediated diseases", GENES AND IMMUNITY, vol. 15, no. 8, 21 August 2014 (2014-08-21), GB, pages 521 - 527, XP055849007, ISSN: 1466-4879, Retrieved from the Internet DOI: 10.1038/gene.2014.46
DIMOPOULOU CHRYSOULA ET AL: "Variant in ERAP1 promoter region is associated with low expression in a patient with a Behçet-like MHC-I-opathy", JOURNAL OF HUMAN GENETICS, NATURE PUBLISHING GROUP, GB; JP, vol. 65, no. 3, 23 December 2019 (2019-12-23), pages 325 - 335, XP036998293, ISSN: 1434-5161, [retrieved on 20191223], DOI: 10.1038/S10038-019-0709-Y
BABAIE FARHAD ET AL: "The roles of ERAP1 and ERAP2 in autoimmunity and cancer immunity: New insights and perspective", MOLECULAR IMMUNOLOGY, PERGAMON, GB, vol. 121, 2 March 2020 (2020-03-02), pages 7 - 19, XP086137735, ISSN: 0161-5890, [retrieved on 20200302], DOI: 10.1016/J.MOLIMM.2020.02.020
ZERVOUDI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 110, 2013, pages 19890 - 19895
MEDVE ET AL., EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 211, 2021, pages 113053
WEGLARZ-TOMCZAK ET AL., BIOORG. MED. CHEM. LETT., vol. 26, 2016, pages 4122 - 4126
GEORGIADIS ET AL., CURR. MEDIC. CHEM., vol. 26, 2019, pages 2715 - 2729
GOTO ET AL., J. IMMUNOL., vol. 192, 2014, pages 4443 - 4452
ZERVOUDI ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 110, 2013, pages 19890 - 19895
WEGLARZ-TOMCZAK ET AL., BIOORG. MED. CHE, LETT., vol. 26, 2016, pages 4122 - 4126
LIDDLE ET AL., J. MED. CHEM., vol. 63, 2020, pages 3348 - 3358
GEORGIADIS ET AL., CUR. MED. CHEM., vol. 26, 2019, pages 2715 - 2729
DEDDOUCHE-GRASS ET AL., ACS MED. CHEM. LETT., vol. 12, 2021, pages 1137 - 1142
LEVERING ET AL., CYTOMETRY B CLIN. CYTOM., vol. 54, 2003, pages 28 - 38
DRABBELS ET AL., BLOOD, vol. 118, 2011, pages e149 - 55
BRAUD ET AL., CURR. BIOL., vol. 8, 1998, pages 1 - 10
SETINI ET AL., HUM. IMMUNOL., vol. 46, 1996, pages 69 - 81
MARGET ET AL., MOL. IMMUNOL., vol. 42, 2005, pages 643 - 649
BAI ET AL., METHODS MOL. BIOL., vol. 1802, 2018, pages 193 - 201
ANDRES ET AL., PLOS GENETICS, vol. 6, 2010, pages 1 - 13
MANIATIS ET AL.: "Molecular Cloning - A Laboratory Manual", 1989, COLD SPRING HARBOR PRESS
VAN HOUT ET AL., NATURE, vol. 586, 2020, pages 749 - 756
REID ET AL., BMC BIOINFORMATICS, vol. 15, 2014, pages 30
BAI ET AL., BMC GENOMICS, vol. 15, 2014, pages 325
ROBINSON ET AL., HUM. IMMUNOL., vol. 77, 2016, pages 233 - 237
JIA ET AL., PLOS ONE, vol. 8, 2013, pages e64683
RICH, ANN.N.Y. ACAD. SCI., vol. 1079, 2006, pages 1 - 8
DELANEAU ET AL., NAT. COMMUN., vol. 10, 2019, pages 5436
DAS ET AL., NAT. GENET., vol. 48, 2016, pages 1284 - 1287
MBATCHOU ET AL., BIORXIV, 2020
ZHOU ET AL., NAT. GENET., vol. 50, 2018, pages 1335 - 1341
PURCELL ET AL., AM. J. HUM. GENET., vol. 81, 2007, pages 559 - 575
MCKENZIE ET AL., GENES IMMUN., vol. 1, 1999, pages 120 - 129
KUIPER ET AL., HUM. MOL. GENET., vol. 23, 2014, pages 6081 - 6087
KUIPER ET AL., HUM. MOL. GENET., vol. 27, 2018, pages 4333 - 4343
PALADINI ET AL., SCI. REP., vol. 8, 2018, pages 10398
ANDRES ET AL., PLOS GENET., vol. 6, 2010, pages el001157
COULOMBE-HUNTINGTON ET AL., PLOS GENET., vol. 5, 2009, pages el000766
MINOS ET AL., ORPHANET J. RARE DIS., vol. 11, 2016, pages 61
SANZ-BRAVO ET AL., MOL. CELL PROTEOMICS, vol. 17, 2018, pages 1308 - 1323
BABAIE ET AL., MOL. IMMUNOL., vol. 121, 2020, pages 7 - 19
YAO ET AL., HUM. IMMUNOL., vol. 80, 2019, pages 325 - 334
EVANS ET AL., NAT. GENET., vol. 43, 2011, pages 761 - 767
WISNIEWSKI ET AL., HUM. IMMUNOL., vol. 79, 2018, pages 109 - 116
NAT. GENET., vol. 42, 2010, pages 985 - 990
TAKEUCHI ET AL., ANN. RHEUM. DIS., vol. 75, 2016, pages 2208 - 2211
GUASP ET AL., J. BIOL. CHEM., vol. 292, 2017, pages 9680 - 9689
MCKENZIE ET AL., GENES IMMUN, vol. 1, 1999, pages 120 - 129
Attorney, Agent or Firm:
LEGAARD, Paul K. (US)
Download PDF:
Claims:
DOCKET NO.: 189238.05102 (3358) (10814W001)

- 165 -

What Is Claimed Is:

1. A method of treating a subject having an immune disorder, the method comprising administering to the subject a therapeutically effective amount of an Endoplasmic Reticulum Aminopeptidase 2 (ERAP2) inhibitor.

2. The method according to claim 1, wherein the immune disorder is an MHC-I- opathy.

3. The method according to claim 2, wherein the MHC-l-opathy is Birdshot Chorioretinopathy (BSCR).

4. The method according to claim 3, the method further comprising detecting the presence or absence of an HLA-Awl9 allele in a biological sample obtained from the subject.

5. The method according to claim 4, the method further comprising determining whether the subject has one or two copies of an HLA-Awl9 allele.

6. The method according to any one of claims 3 to 5, wherein the subject is HLA- Awl9+.

7. The method according to claim 6, wherein the subject is HLA-A*29+, HLA-A*30+, HLA-A*31+, or HLA-A*33+, or any combination thereof.

8. The method according to claim 6 or claim 7 , wherein the subject has two copies of HLA-Awl9.

9. The method according to claim 8, wherein the subject is HLA-A*29+/HLA-A*30+.

10. The method according to claim 8, wherein the subject is HLA-A*29+/HLA-A*31+.

11. The method according to claim 8, wherein the subject is HLA-A*29+/HLA-A*33+.

12. The method according to any one of claims 3 to 11, the method further comprising administering to the subject an HLA-Awl9 inhibitor.

13. The method according to claim 12, wherein the HLA-Awl9 inhibitor is an antibody.

14. The method according to claim 13, wherein the antibody is an anti-HLA-A*29 antibody.

15. The method according to claim 12, wherein the HLA-Awl9 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 166 -

16. The method according to claim 15, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-Awl9.

17. The method according to claim 16, wherein the HLA-Awl9 is HLA-A*29.

18. The method according to claim 2, wherein the MHC-l-opathy is Ankylosing Spondylitis (AS).

19. The method according to claim 18, the method further comprising detecting the presence or absence of HLA-B*27 or HLA-B*40 in a biological sample obtained from the subject.

20. The method according to claim 19, the method further comprising determining whether the subject has one or two copies of HLA-B*27 or HLA-B*40.

21. The method according to any one of claims 18 to 20, wherein the subject is HLA- B*27+ or HLA-B*40+.

22. The method according to claim 21, wherein the subject has two copies of HLA-B*27 or HLA-B*40.

23. The method according to any one of claims 18 to 22, the method further comprising administering to the subject an HLA-B*27 inhibitor or an HLA-B*40 inhibitor.

24. The method according to claim 23, wherein the HLA-B*27 inhibitor or HLA-B*40 inhibitor is an antibody.

25. The method according to claim 24, wherein the antibody is an anti-HLA-B*27 antibody or an anti-HLA-B*40 antibody.

26. The method according to claim 23, wherein the HLA-B*27 inhibitor or HLA-B*40 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

27. The method according to claim 26, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*27 or an HLA-B*40.

28. The method according to claim 2, wherein the MHC-l-opathy is Behget's disease.

29. The method according to claim 28, the method further comprising detecting the presence or absence of HLA-B*51 in a biological sample obtained from the subject. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 167 -

30. The method according to claim 29, the method further comprising determining whether the subject has one or two copies of HLA-B*51.

31. The method according to any one of claims 28 to 30, wherein the subject is HLA- B*51+.

32. The method according to claim 31, wherein the subject has two copies of HLA- B*51.

33. The method according to any one of claims 28 to 32, the method further comprising administering to the subject an HLA-B*51 inhibitor.

34. The method according to claim 33, wherein the HLA-B*51 inhibitor is an antibody.

35. The method according to claim 34, wherein the antibody is an anti-HLA-B*51 antibody.

36. The method according to claim 33, wherein the HLA-B*51 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

37. The method according to claim 36, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*51.

38. The method according to claim 2, wherein the MHC-l-opathy is psoriasis.

39. The method according to claim 38, the method further comprising detecting the presence or absence of HLA-C*06 in a biological sample obtained from the subject.

40. The method according to claim 39, the method further comprising determining whether the subject has one or two copies of HLA-C*06.

41. The method according to any one of claims 38 to 40, wherein the subject is HLA- C*06+.

42. The method according to claim 41, wherein the subject has two copies of HLA- C*06.

43. The method according to any one of claims 38 to 42, the method further comprising administering to the subject an HLA-C*06 inhibitor.

44. The method according to claim 43, wherein the HLA-C*06 inhibitor is an antibody.

45. The method according to claim 44, wherein the antibody is an anti-HLA-C*06 antibody. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 168 -

46. The method according to claim 43, wherein the HLA-C*06 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

47. The method according to claim 46, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-C*06.

48. The method according to claim 2, wherein the MHC-l-opathy is Juvenile Idiopathic Arthritis (JIA).

49. The method according to claim 48, the method further comprising detecting the presence or absence of HLA-B*27 in a biological sample obtained from the subject.

50. The method according to claim 49, the method further comprising determining whether the subject has one or two copies of HLA-B*27.

51. The method according to any one of claims 48 to 50, wherein the subject is HLA- B*27+.

52. The method according to claim 51, wherein the subject has two copies of HLA- B*27.

53. The method according to any one of claims 48 to 52, the method further comprising administering to the subject an HLA-B*27 inhibitor.

54. The method according to claim 53, wherein the HLA-B*27 inhibitor is an antibody.

55. The method according to claim 54, wherein the antibody is an anti-HLA-B*27 antibody.

56. The method according to claim 53, wherein the HLA-B*27 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

57. The method according to claim 56, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*27.

58. The method according to claim 2, wherein the MHC-l-opathy is inflammatory bowel disease (IBD) or Crohn's disease (CD).

59. The method according to claim 58, the method further comprising detecting the presence or absence of HLA-C*07 in a biological sample obtained from the subject. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 169 -

60. The method according to claim 59, the method further comprising determining whether the subject has one or two copies of HLA-C*07.

61. The method according to any one of claims 58 to 60, wherein the subject is HLA- C*07+.

62. The method according to claim 61, wherein the subject has two copies of HLA- C*07.

63. The method according to any one of claims 58 to 62, the method further comprising administering to the subject an HLA-C*07 inhibitor.

64. The method according to claim 63, wherein the HLA-C*07 inhibitor is an antibody.

65. The method according to claim 64, wherein the antibody is an anti-HLA-C*07 antibody.

66. The method according to claim 63, wherein the HLA-C*07 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

67. The method according to claim 66, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-C*07.

68. The method according to any one of claims 1 to 67, wherein the ERAP2 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

69. The method according to claim 68, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to ERAP2 mRNA.

70. The method according to any one of claims 1 to 67, wherein the ERAP2 inhibitor comprises an anti-ERAP2 antibody.

71. The method according to any one of claims 1 to 67, wherein the ERAP2 inhibitor comprises a pseudopeptide.

72. The method according to claim 71, wherein the pseudopeptide is a phosphinic pseudopeptide.

73. The method according to claim 72, wherein the phosphinic pseudopeptide is DG002 or DG013. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 170 -

74. The method according to any one of claims 1 to 67, wherein the ERAP2 inhibitor comprises a small molecule.

75. The method according to any one of claims 1 to 74, further comprising administering to the subject a therapeutically effective amount of an Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) agonist or inhibitor.

76. The method according to claim 75, wherein the ERAP1 agonist comprises an oligonucleotide.

77. The method according to claim 76, wherein the oligonucleotide is ODN1826.

78. The method according to claim 75, wherein the ERAP1 agonist comprises a lipopeptide.

79. The method according to claim 78, wherein the lipopeptide is Pam3CSK4 or FSL-1.

80. The method according to claim 75, wherein the ERAP1 agonist comprises a small molecule.

81. The method according to claim 75, wherein the ERAP1 inhibitor comprises an inhibitory nucleic acid molecule.

82. The method according to claim 75, wherein the ERAP1 inhibitor comprises a small molecule.

83. A method of treating a subject having an MHC-l-opathy, the method comprising: performing or having performed an assay on a biological sample from the subject to determine whether the subject comprises: i) an MHC-l-opathy-related HLA genotype; and ii) a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein; and administering to the subject a therapeutically effective amount of an ERAP2 inhibitor, wherein the subject comprises both an MHC-l-opathy-related HLA genotype and a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein; wherein the presence of both the MHC-l-opathy-related HLA genotype and the functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein indicates that the subject is a candidate for treating the MHC-l-opathy by inhibiting ERAP2. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 171 -

84. The method according to claim 83, wherein the MHC-l-opathy is Birdshot Chorioretinopathy (BSCR) and the MHC-l-opathy-related HLA genotype comprises an HLA- Awl9 allele.

85. The method according to claim 84, wherein HLA-Awl9 allele is an HLA-A*29 allele, an HLA-A*30 allele, an HLA-A*31 allele, or an HLA-A*33 allele, or any combination thereof.

86. The method according to claim 84 or claim 85, wherein the subject has two copies of the HLA-Awl9 allele.

87. The method according to claim 86, wherein the subject is HLA-A*29+/HLA-A*30+.

88. The method according to claim 86, wherein the subject is HLA-A*29+/HLA-A*31+.

89. The method according to claim 86, wherein the subject is HLA-A*29+/HLA-A*33+.

90. The method according to any one of claims 84 to 89, the method further comprising administering to the subject an HLA-Awl9 inhibitor.

91. The method according to claim 90, wherein the HLA-Awl9 inhibitor is an antibody.

92. The method according to claim 91, wherein the antibody is an anti-HLA-A*29 antibody.

93. The method according to claim 90, wherein the HLA-Awl9 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

94. The method according to claim 93, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-Awl9.

95. The method according to claim 94, wherein the HLA-Awl9 is HLA-A*29.

96. The method according to claim 83, wherein the MHC-l-opathy is Ankylosing

Spondylitis (AS) and the MHC-l-opathy-related HLA genotype comprises an HLA-B*27 allele or an HLA-B*40 allele.

97. The method according to claim 96, wherein the subject has two copies of HLA-B*27 or HLA-B*40.

98. The method according to claim 96 or claim 97, the method further comprising administering to the subject an HLA-B*27 inhibitor or an HLA-B*40 inhibitor.

99. The method according to claim 96, wherein the HLA-B*27 inhibitor or HLA-B*40 inhibitor is an antibody. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 172 -

100. The method according to claim 99, wherein the antibody is an anti-HLA-B*27 antibody or an anti-HLA-B*40 antibody.

101. The method according to claim 98, wherein the HLA-B*27 inhibitor or HLA-B*40 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

102. The method according to claim 101, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*27 or an HLA-B*40.

103. The method according to claim 83, wherein the MHC-l-opathy is Behget's disease and the MHC-l-opathy-related HLA genotype comprises an HLA-B*51 allele.

104. The method according to claim 103, wherein the subject has two copies of HLA- B*51.

105. The method according to claim 103 or claim 104, the method further comprising administering to the subject an HLA-B*51 inhibitor.

106. The method according to claim 105, wherein the HLA-B*51 inhibitor is an antibody.

107. The method according to claim 106, wherein the antibody is an anti-HLA-B*51 antibody.

108. The method according to claim 105, wherein the HLA-B*51 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

109. The method according to claim 108, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*51.

110. The method according to claim 83, wherein the MHC-l-opathy is psoriasis and the MHC-l-opathy-related HLA genotype comprises an HLA-C*06 allele.

111. The method according to claim 110, wherein the subject has two copies of HLA- C*06.

112. The method according to claim 110 or claim 111, the method further comprising administering to the subject an HLA-C*06 inhibitor.

113. The method according to claim 112, wherein the HLA-C*06 inhibitor is an antibody.

114. The method according to claim 113, wherein the antibody is an anti-HLA-C*06 antibody. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 173 -

115. The method according to claim 112, wherein the HLA-C*06 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

116. The method according to claim 115, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-C*06.

117. The method according to claim 83, wherein the MHC-l-opathy is Juvenile Idiopathic Arthritis (JIA) and the MHC-l-opathy-related HLA genotype comprises an HLA-B*27.

118. The method according to claim 117, wherein the subject has two copies of HLA- B*27.

119. The method according to claim 117 or claim 118, the method further comprising administering to the subject an HLA-B*27 inhibitor.

120. The method according to claim 119, wherein the HLA-B*27 inhibitor is an antibody.

121. The method according to claim 120, wherein the antibody is an anti-HLA-B*27 antibody.

122. The method according to claim 119, wherein the HLA-B*27 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

123. The method according to claim 122, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*27.

124. The method according to claim 83, wherein the MHC-l-opathy is inflammatory bowel disease (IBD) or Crohn's disease (CD) and the MHC-l-opathy-related HLA genotype comprises an HLA-C*07 allele.

125. The method according to claim 124, wherein the subject has two copies of HLA- C*07.

126. The method according to claim 124 or claim 125, the method further comprising administering to the subject an HLA-C*07 inhibitor.

127. The method according to claim 126, wherein the HLA-C*07 inhibitor is an antibody.

128. The method according to claim 127, wherein the antibody is an anti-HLA-C*07 antibody. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 174 -

129. The method according to claim 126, wherein the HLA-C*07 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

130. The method according to claim 129, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-C*07.

131. The method according to any one of claims 83 to 130, wherein the nucleic acid molecule comprises genomic DNA, mRNA, or cDNA obtained from mRNA.

132. The method according to any one of claims 83 to 131, wherein the ERAP2 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

133. The method according to claim 132, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to ERAP2 mRNA.

134. The method according to any one of claims 83 to 131, wherein the ERAP2 inhibitor comprises an anti-ERAP2 antibody.

135. The method according to any one of claims 83 to 131, wherein the ERAP2 inhibitor comprises a pseudopeptide.

136. The method according to claim 135, wherein the pseudopeptide is a phosphinic pseudopeptide.

137. The method according to claim 136, wherein the phosphinic pseudopeptide is DG002 or DG013.

138. The method according to any one of claims 83 to 131, wherein the ERAP2 inhibitor comprises a small molecule.

139. The method according to any one of claims 83 to 138, wherein: the assay performed or having been performed on the biological sample from the subject further determines whether the subject comprises a functional Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) protein or a nucleic acid molecule encoding a functional ERAP1 protein; and the method further comprises administering to the subject a therapeutically effective amount of an ERAP1 agonist, wherein the subject comprises an MHC-l-opathy- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 175 - related HLA genotype and does not comprise a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein; wherein the presence of an MHC-l-opathy-related HLA genotype and the absence of a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein indicates that the subject is a candidate for treating the MHC-l-opathy by activating ERAP1.

140. The method according to claim 139, wherein the ERAP1 agonist comprises a comprises an oligonucleotide.

141. The method according to claim 140, wherein the oligonucleotide is ODN1826.

142. The method according to claim 139, wherein the ERAP1 agonist comprises a lipopeptide.

143. The method according to claim 142, wherein the lipopeptide is Pam3CSK4 or F5L-1.

144. The method according to claim 139, wherein the ERAP1 agonist comprises a small molecule.

145. The method according to any one of claims 83 to 138, wherein: the assay performed or having been performed on the biological sample from the subject further determines whether the subject comprises a functional Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) protein or a nucleic acid molecule encoding a functional ERAP1 protein; and the method further comprises administering to the subject a therapeutically effective amount of an ERAP1 inhibitor, wherein the subject comprises an MHC-l-opathy- related HLA genotype and comprises a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein; wherein the presence of an MHC-l-opathy-related HLA genotype and the presence of a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein indicates that the subject is a candidate for treating the MHC-l-opathy by inhibiting ERAP1.

146. The method according to claim 145, wherein the ERAP1 inhibitor comprises an oligonucleotide. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 176 -

147. The method according to claim 145, wherein the ERAP1 inhibitor comprises a small molecule.

148. A method of identifying a subject having an increased risk for developing an MHC-I- opathy, the method comprising: performing or having performed an assay on a biological sample from the subject to determine whether the subject comprises: i) an MHC-l-opathy-related HLA genotype; and ii) a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein; wherein: when the subject has both the MHC-l-opathy-related HLA genotype and the functional ERAP2 protein or the nucleic acid molecule encoding the functional ERAP2 protein, then the subject has an increased risk of developing the MHC-l-opathy; when the subject lacks the MHC-l-opathy-related HLA genotype, or lacks the functional ERAP2 protein or the nucleic acid molecule encoding the functional ERAP2 protein, or lacks both, then the subject has a decreased risk of developing the MHC-l- opathy; and when the subject comprises two copies of the MHC-l-opathy-related HLA genotype, then the subject has an increased risk of developing the MHC-l-opathy compared to comprising a single copy of the MHC-l-opathy-related HLA genotype.

149. The method according to claim 148, wherein the MHC-l-opathy is Birdshot Chorioretinopathy (BSCR) and the MHC-l-opathy-related HLA genotype comprises an HLA- Awl9 allele.

150. The method according to claim 149, wherein HLA-Awl9 allele is an HLA-A*29 allele, an HLA-A*30 allele, an HLA-A*31 allele, or an HLA-A*33 allele, or any combination thereof.

151. The method according to claim 149 or claim 150, wherein the subject has two copies of the HLA-Awl9 allele.

152. The method according to claim 151, wherein the subject is HLA-A*29+/HLA-A*30+.

153. The method according to claim 151, wherein the subject is HLA-A*29+/HLA-A*31+.

154. The method according to claim 151, wherein the subject is HLA-A*29+/HLA-A*33+. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 177 -

155. The method according to any one of claims 149 to 154, the method further comprising administering an HLA-Awl9 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

156. The method according to claim 155, wherein the HLA-Awl9 inhibitor is an antibody.

157. The method according to claim 156, wherein the antibody is an anti-HLA-A*29 antibody.

158. The method according to claim 155, wherein the HLA-Awl9 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

159. The method according to claim 158, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-Awl9.

160. The method according to claim 159, wherein the HLA-Awl9 is HLA-A*29.

161. The method according to claim 148, wherein the MHC-l-opathy is Ankylosing Spondylitis (AS) and the MHC-l-opathy-related HLA genotype comprises an HLA-B*27 allele or an HLA-B*40 allele.

162. The method according to claim 161, wherein the subject has two copies of HLA- B*27 or HLA-B*40.

163. The method according to claim 161 or claim 162, the method further comprising administering an HLA-B*27 inhibitor or an HLA-B*40 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

164. The method according to claim 163, wherein the HLA-B*27 inhibitor or HLA-B*40 inhibitor is an antibody.

165. The method according to claim 164, wherein the antibody is an anti-HLA-B*27 antibody or an anti-HLA-B*40 antibody.

166. The method according to claim 163, wherein the HLA-B*27 inhibitor or HLA-B*40 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

167. The method according to claim 166, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*27 or an HLA-B*40. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 178 -

168. The method according to claim 148, wherein the MHC-l-opathy is Behget's disease and the MHC-l-opathy-related HLA genotype comprises an HLA-B*51 allele.

169. The method according to claim 168, wherein the subject has two copies of HLA- B*51.

170. The method according to claim 168 or claim 169, the method further comprising administering an HLA-B*51 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

171. The method according to claim 170, wherein the HLA-B*51 inhibitor is an antibody.

172. The method according to claim 170, wherein the antibody is an anti-HLA-B*51 antibody.

173. The method according to claim 170, wherein the HLA-B*51 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

174. The method according to claim 173, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*51.

175. The method according to claim 148, wherein the MHC-l-opathy is psoriasis and the MHC-l-opathy-related HLA genotype comprises an HLA-C*06 allele.

176. The method according to claim 175, wherein the subject has two copies of HLA- C*06.

177. The method according to claim 175 or claim 176, the method further comprising administering an HLA-C*06 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

178. The method according to claim 177, wherein the HLA-C*06 inhibitor is an antibody.

179. The method according to claim 178, wherein the antibody is an anti-HLA-C*06 antibody.

180. The method according to claim 177, wherein the HLA-C*06 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

181. The method according to claim 180, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-C*06. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 179 -

182. The method according to claim 148, wherein the MHC-l-opathy is Juvenile Idiopathic Arthritis (JIA) and the MHC-l-opathy-related HLA genotype comprises an HLA- B*27.

183. The method according to claim 182, wherein the subject has two copies of HLA- B*27.

184. The method according to claim 182 or claim 183, the method further comprising administering an HLA-B*27 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

185. The method according to claim 184, wherein the HLA-B*27 inhibitor is an antibody.

186. The method according to claim 185, wherein the antibody is an anti-HLA-B*27 antibody.

187. The method according to claim 184, wherein the HLA-B*27 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

188. The method according to claim 187, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-B*27.

189. The method according to claim 148, wherein the MHC-l-opathy is inflammatory bowel disease (IBD) or Crohn's disease (CD) and the MHC-l-opathy-related HLA genotype comprises an HLA-C*07 allele.

190. The method according to claim 189, wherein the subject has two copies of HLA- C*07.

191. The method according to claim 189 or claim 190, the method further comprising administering an HLA-C*07 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

192. The method according to claim 191, wherein the HLA-C*07 inhibitor is an antibody.

193. The method according to claim 192, wherein the antibody is an anti-HLA-C*07 antibody.

194. The method according to claim 191, wherein the HLA-C*07 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 180 -

195. The method according to claim 194, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to an HLA-C*07.

196. The method according to any one of claims 148 to 195, wherein the nucleic acid molecule comprises genomic DNA, mRNA, or cDNA obtained from mRNA.

197. The method according to any one of claims 148 to 196, the method further comprising administering an ERAP2 inhibitor to the subject having an increased risk of developing the MHC-l-opathy.

198. The method according to claim 197, wherein the ERAP2 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

199. The method according to claim 198, wherein the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA that hybridizes to ERAP2 mRNA.

200. The method according to claim 197, wherein the ERAP2 inhibitor comprises an anti- ERAP2 antibody.

201. The method according to claim 197, wherein the ERAP2 inhibitor comprises a pseudopeptide.

202. The method according to claim 201, wherein the pseudopeptide is a phosphinic pseudopeptide.

203. The method according to claim 202, wherein the phosphinic pseudopeptide is DG002 or DG013.

204. The method according to claim 197, wherein the ERAP2 inhibitor comprises a small molecule.

205. The method according to any one of claims 148 to 204, wherein: the assay performed or having been performed on the biological sample from the subject further determines whether the subject comprises a functional Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) protein or a nucleic acid molecule encoding a functional ERAP1 protein; and DOCKET NO.: 189238.05102 (3358) (10814W001)

- 181 - when the subject has the MHC-l-opathy-related HLA genotype and lacks the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, then the subject has an increased risk of developing the MHC-l-opathy; and when the subject lacks the MHC-l-opathy-related HLA genotype, or has the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, or both, then the subject has a decreased risk of developing the MHC-l-opathy.

206. The method according to claim 205, the method further comprising administering an ERAP1 agonist to the subject having an increased risk of developing the MHC-l-opathy.

207. The method according to claim 206, wherein the ERAP1 agonist comprises a comprises an oligonucleotide.

208. The method according to claim 207, wherein the oligonucleotide is ODN1826.

209. The method according to claim 206, wherein the ERAP1 agonist comprises a lipopeptide.

210. The method according to claim 209, wherein the lipopeptide is Pam3CSK4 or FSL-1

211. The method according to claim 206, wherein the ERAP1 agonist comprises a small molecule.

212. The method according to any one of claims 148 to 204, wherein: the assay performed or having been performed on the biological sample from the subject further determines whether the subject comprises a functional Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) protein or a nucleic acid molecule encoding a functional ERAP1 protein; and when the subject has the MHC-l-opathy-related HLA genotype and has the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, then the subject has an increased risk of developing the MHC-l-opathy; and when the subject lacks the MHC-l-opathy-related HLA genotype, or does not comprise the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, or both, then the subject has a decreased risk of developing the MHC-l- opathy.

213. The method according to claim 212, the method further comprising administering an ERAP1 inhibitor to the subject having an increased risk of developing the MHC-l-opathy. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 182 -

214. The method according to claim 213, wherein the ERAP1 inhibitor comprises an inhibitory nucleic acid molecule.

215. The method according to claim 213, wherein the ERAP1 inhibitor comprises a small molecule.

Description:
Modulation Of Endoplasmic Reticulum Aminopeptidase 2 (ERAP2)-Mediated Immune Response

Reference To Sequence Listing

This application includes a Sequence Listing filed electronically as a text file named 18923805102SEQ, created on July 23, 2021, with a size of 1,243 kilobytes. The Sequence Listing is incorporated herein by reference.

Field

The present disclosure is directed, in part, to methods of treating subjects having an immune disorder by administering a therapeutically effective amount of an Endoplasmic Reticulum Aminopeptidase 2 (ERAP2) inhibitor to the subject, and optionally an Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) agonist or inhibitor and/or an HLA-Awl9 inhibitor, and also provides methods of identifying subjects having an increased risk for developing an MHC-I- opathy.

Background

The cellular immune response in humans relies at least partly on the presentation of small peptides that are 8 to 10 amino acids long, which are bound proteins of the major histocompatibility complex (MHC) (i.e., class I MHC molecules). These small peptides are derived from the proteolytic degradation of proteins (foreign antigens and self-antigens). One source of these antigens come from infected or malignantly transformed cells that express particular protein molecules that, upon degradation, yield distinct antigenic peptides that are presented on the cell surface complexed with MHC class I molecules (MHCI). Cytotoxic T cells can recognize these complexes of MHC molecules with degraded protein antigens and induce apoptotic cell death. Aberrant generation of antigenic peptides can lead to immune system evasion or to autoimmune reactions.

Although most antigenic peptides are initially produced by the proteasome, many of them are larger than the final antigenic epitope and contain one or more additional amino acids at their N-termini. These antigenic peptide precursors are transported into the endoplasmic reticulum (ER), where they are further degraded by at least two different aminopeptidases, ERAP1 and ERAP2, to generate the mature antigenic peptides for complexing with MHC class I molecules. Thus, the activity of ERAP1 and ERAP2 can directly affect the presentation of antigenic peptides complexed with particular MHC molecules in a beneficial or adverse manner, thus altering the immune response. Accordingly, there continues to be a need for identifying subjects that have particular MHC-l-opathies related to ERAP2 activity and treatment of the same.

Birdshot Chorioretinopathy (BSCR) is a rare autoimmune uveitis predominately affecting individuals over the age of 50 of European descent and treated with immunomodulatory therapies. The disease presents with vitritis and gradual decline in vision due to choroidal and retinal inflammatory lesions and atrophy. T cells have been identified in the retinal and choroidal tissues as well as the vitreous of affected BSCR eyes.

Summary

The present disclosure provides methods of treating a subject having an immune disorder, the methods comprising administering to the subject a therapeutically effective amount of an ERAP2 inhibitor. Optionally, an ERAP1 agonist or inhibitor and/or an HLA-Awl9 inhibitor is also administered to the subject. The immune disorder can be an MHC-l-opathy or an MHC-ll-opathy. The MHC-l-opathy can be BSCR, Ankylosing Spondylitis (AS), Behget's disease, psoriasis, Juvenile Idiopathic Arthritis (J I A), inflammatory bowel disease (IBD), or Crohn's disease (CD).

The present disclosure also provides methods of treating a subject having an MHC-l- opathy, the methods comprising: performing or having performed an assay on a biological sample from the subject to determine whether the subject comprises: i) an MHC-l-opathy- related HLA genotype; and ii) a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein; and administering to the subject a therapeutically effective amount of an ERAP2 inhibitor, wherein the subject comprises both an MHC-l-opathy-related HLA genotype and a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein; wherein the presence of both the MHC-l-opathy-related HLA genotype and the functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein indicates that the subject is a candidate for treating the MHC-l-opathy by inhibiting ERAP2. The assay performed or having been performed on the biological sample from the subject can further determine whether the subject comprises a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein; and the methods can further comprise administering to the subject a therapeutically effective amount of an ERAP1 agonist or inhibitor, wherein the subject comprises an MHC-l-opathy-related HLA genotype and does or does not comprise a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein; wherein the presence of an MHC-l-opathy-related HLA genotype and the absence of a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein indicates that the subject is a candidate for treating the MHC-l-opathy by activating ERAP1; wherein the presence of an MHC-l-opathy-related HLA genotype and the presence of a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein indicates that the subject is a candidate for treating the MHC-l-opathy by inhibiting ERAP1.

The present disclosure also provides methods of identifying a subject having an increased risk for developing an MHC-l-opathy, the methods comprising: performing or having performed an assay on a biological sample from the subject to determine whether the subject comprises: i) an MHC-l-opathy-related HLA genotype; and ii) a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein; wherein: when the subject has both the MHC-l-opathy-related HLA genotype and the functional ERAP2 protein or the nucleic acid molecule encoding the functional ERAP2 protein, then the subject has an increased risk of developing the MHC-l-opathy; when the subject lacks the MHC-l-opathy-related HLA genotype, or lacks the functional ERAP2 protein or the nucleic acid molecule encoding the functional ERAP2 protein, or lacks both, then the subject has a decreased risk of developing the MHC-l- opathy; and when the subject comprises two copies of the MHC-l-opathy-related HLA genotype, then the subject has an increased risk of developing the MHC-l-opathy compared to comprising a single copy of the MHC-l-opathy-related HLA genotype. The assay performed or having been performed on the biological sample from the subject can further determine whether the subject comprises a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein. When the subject has the MHC-l-opathy-related HLA genotype and lacks the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, then the subject has an increased risk of developing the MHC-l-opathy; and when the subject lacks the MHC-l-opathy-related HLA genotype, or has the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, or both, then the subject has a decreased risk of developing the MHC-l-opathy. Brief Description Of The Drawings

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

Figure 1 shows a table of HLA-A 2nd allele frequencies in the French cohort compared to UKB and G HS EUR A29 carriers. Alleles belonging to the Awl9 broad antigen group that increase risk are A29, A30, A31 and A33 (red) and A32 exhibits protection (green). A Fisher's exact test combining all Awl9 risk alleles presents the strongest enrichment in all comparisons. Only alleles that have three or more case carriers are presented. Table is sorted by p-values when comparing case frequencies against A29 controls in UKB.

Figure 2 shows Awl9 enrichment in Birdshot cases. Odds-ratio for BSCR, comparing frequencies of 14 FILA-A alleles that are present in three or more cases (>1%, x-axis) in 286 UParis cases compared with 108 UParis controls (Blue), GHS control cohort #1 (n= 4,014, dark green), GHS control cohort #2 (n= 2,829, bright green) and UKB controls (n= 38,543, yellow). Awl9 alleles show the highest ORs (red box) that replicates with large A29 control cohorts, with the exception of A32 that is depleted in cases (green box). * p<0.01

Figure 3 shows a table of HLA-A 2nd allele frequencies in the French cohort compared to UKB and GHS EUR A29 carriers. Alleles belonging to the Awl9 broad antigen group that increase risk are A29, A30, A31 and A33 (red) and A32 exhibits protection (green). A logistic regression test with covariates included for sex and six principal components, calculated based on genetic array data for each analytic set. Results are presented for all HLA-A alleles that have three or more case carriers. Alleles are sorted as in Figure 1. *Three principal components.

Figure 4 shows a table of haplotype analysis of ERAP1. Haplotype analysis examining the association of all eight ERAP1 haplotypes with the case-control status, showing that Hapl and Hap2 are strongly associated with protection from BSCR.

Figure 5 shows a table of top SNPs in ERAP1 and ERAP2 regions. Variants in ERAP1 and ERAP2 are genome-wide significant when analyzed together with previous results (125 cases and 670 controls (Kuiper 2014). Rsl0044354 is the top association in the ERAP1-ERAP2 locus in the previous GWAS of Dutch and Spanish cohorts, while rs27432 is the top association in the region in the current French cohort. The LD between the two loci is also presented. *The reference A-allele is the minor allele, risk is the G-allele. Figure 6 shows ERAP2 splice region variant is protective for BSCR. The common ERAP2 splice region variant rs2248374 that disrupts ERAP2 expression is protective in the current BSCR cohort and the previous Spanish and Dutch cohorts.

Figure 7 shows the combined risk of ERAP1, ERAP2 and two copies of Awl9. Utilizing 286 Birdshot cases and 4,014 controls from G HS cohort #1 to calculate additive risk while combining risk factors in ERAP1, ERAP2 and Awl9. Panel A) An additive genotype model of ERAP2 risk signal tagged by rsl0044354 and single (A29/-) or double (A29/Awl9) Awl9 copies relative to lowest risk combination of rsl0044354-CC and one copy of Awl9 allele (A29). Panel B) An additive genotype model of ERAP1 risk signal tagged by rs27432 and single (A29/-) or double (A29/Awl9) Awl9 copies relative to lowest risk combination of rs27432 -AA and one copy of Awl9 allele (A29). Panel C) An additive genotype model of ERAP1 risk signal tagged by rs27432 and ERAP2 signal tagged by rsl0044354 relative to lowest risk combination of rs27432- AA and rsl0044354-CC. Panel D) An additive genotype model of ERAP1 and ERAP2 risk signals and single (A29/-) or double (A29/Awl9) Awl9 copies relative to lowest risk combination. The genotypes are combined as following: 0 = ERAP1 and ERAP2 homozygous for protective allele. 1/[01],[01]/1 = either homozygous protective or heterozygous genotypes of both ERAP1 and ERAP2. 2/.,./2 = homozygous risk allele of either ERAP1 or ERAP2. 2/2 = homozygous risk allele of both ERAP1 and ERAP2.

Figure 8 shows a table of the combined risk of ERAP2 and Awl9. Utilizing 286 Birdshot cases and 4,014 controls from G HS cohort #1 to calculate additive risk while combining risk factors in ERAP2 and Awl9. An additive genotype model of ERAP2 risk signal tagged by rsl0044354 and single (A29/-) or double (A29/Awl9) Awl9 copies relative to lowest risk combination of rsl0044354-CC and one copy of Awl9 allele (A29).

Figure 9 shows a table of the combined risk of ERAP1 and Awl9. Utilizing 286 Birdshot cases and 4,014 controls from G HS cohort #1 to calculate additive risk while combining risk factors in ERAP1 and Awl9. An additive genotype model of ERAP1 risk signal tagged by rs27432 and single (A29/-) or double (A29/Awl9) Awl9 copies relative to lowest risk combination of rs27432-AA and one copy of Awl9 allele (A29).

Figure 10 shows a table of the combined risk of ERAP1 and ERAP2. Utilizing 286 Birdshot cases and 4,014 controls from GHS cohort #1 to calculate additive risk while combining risk factors in ERAP1 and ERAP2. An additive genotype model of ERAP1 risk signal tagged by rs27432 and ERAP2 signal tagged by rsl0044354 relative to lowest risk combination of rs27432- AA and rsl0044354-CC.

Figure 11 shows a table of the combined risk of ERAP1, ERAP2 and Awl9. Utilizing 286 Birdshot cases and 4,014 controls from GHS cohort #1 to calculate additive risk while combining risk factors in ERAP1, ERAP2 and Awl9. An additive genotype model of ERAP1 and ERAP2 risk signals and single (A29/-) or double (A29/Awl9) Awl9 copies relative to lowest risk combination. The genotypes are combined as following: 0 = ERAP1 and ERAP2 homozygous for protective allele. 1/[01],[01]/1 = either homozygous protective or heterozygous genotypes of both ERAP1 and ERAP2. 2/.,./2 = homozygous risk allele of either ERAP1 or ERAP2. 2/2 = homozygous risk allele of both ERAP1 and ERAP2.

Figure 12 shows differences between risk Awl9 alleles and A32. Panel A) Sequence differences between risk Awl9 alleles (red) and protective A32 allele (green). A32 exhibits F at position 9 as is the reference A:01:01 allele, while risk alleles are either T or S at that position. The Bw4 epitope sequence is apparent at positions 79-83 or A32 only.

Description Of Embodiments

Various terms relating to aspects of the present disclosure are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art, unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein.

Unless otherwise expressly stated, it is in no way intended that any method or aspect set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not specifically state in the claims or descriptions that the steps are to be limited to a specific order, it is in no way intended that an order be inferred, in any respect. This holds for any possible non-expressed basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, or the number or type of aspects described in the specification.

As used herein, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise.

As used herein, the term "about" means that the recited numerical value is approximate and small variations would not significantly affect the practice of the disclosed embodiments. Where a numerical value is used, unless indicated otherwise by the context, the term "about" means the numerical value can vary by ±10% and remain within the scope of the disclosed embodiments.

As used herein, the term "comprising" may be replaced with "consisting" or "consisting essentially of" in particular embodiments as desired.

As used herein, the terms "nucleic acid", "nucleic acid molecule", "nucleic acid sequence", "polynucleotide", or "oligonucleotide" can comprise a polymeric form of nucleotides of any length, can comprise DNA and/or RNA, and can be single-stranded, double- stranded, or multiple stranded. One strand of a nucleic acid also refers to its complement.

As used herein, the term "subject" includes any animal, including mammals. Mammals include, but are not limited to, farm animals (such as, for example, horse, cow, pig), companion animals (such as, for example, dog, cat), laboratory animals (such as, for example, mouse, rat, rabbits), and non-human primates (such as, for example, apes and monkeys). In some embodiments, the subject is a human. In some embodiments, the subject is a patient under the care of a physician.

The present disclosure provides methods of treating a subject having an immune disorder, the methods comprising administering to the subject an ERAP2 inhibitor. In some embodiments, the immune disorder is an MHC-l-opathy. In some embodiments, the immune disorder is an MHC-ll-opathy. In some embodiments, the MHC-l-opathy is Birdshot Chorioretinopathy (BSCR), Ankylosing Spondylitis (AS), Behget's disease, psoriasis, Juvenile Idiopathic Arthritis (J I A), inflammatory bowel disease (IBD), or Crohn's disease (CD). In some embodiments, the MHC-l-opathy is BSCR. In some embodiments, the MHC-l-opathy is AS. In some embodiments, the MHC-l-opathy is Behget's disease. In some embodiments, the MHC-l- opathy is psoriasis. In some embodiments, the MHC-l-opathy is JIA. In some embodiments, the MHC-l-opathy is IBD. In some embodiments, the MHC-l-opathy is CD.

In some embodiments, the MHC-l-opathy is BSCR. In some embodiments, the method further comprises detecting the presence or absence of an HLA-Awl9 allele in a biological sample obtained from the subject. In some embodiments, the subject is HLA-Awl9 + . In some embodiments, the subject is or is suspected of being HLA-A*29 + , HLA-A*30 + , HLA-A*31 + , or HLA-A*33 + , or any combination thereof. In some embodiments, the method further comprises determining whether the subject has one or two copies of an HLA-Awl9 allele. In some embodiments, the subject has a single copy of HLA-Awl9. In some embodiments, the subject has two copies of HLA-Awl9. In some embodiments, the subject is HLA-A*29 + /HLA-A*30 + . In some embodiments, the subject is HLA-A*29 + /HLA-A*31 + . In some embodiments, the subject is H LA-A* 29 + /H LA-A*33 + .

In some embodiments, the subject having BSCR is not HLA-A*29 + .

In some embodiments, the subject having BSCR has a copy of at least any two of HLA- A*29, HLA-A*30, HLA-A*31, or HLA-A*33. In some embodiments, the subject having BSCR has a copy of at least any three of HLA-A*29, HLA-A*30, HLA-A*31, or HLA-A*33. In some embodiments, the subject having BSCR has a copy of all of HLA-A*29, HLA-A*30, HLA-A*31, or HLA-A*33.

In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*30. In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*31. In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*33. In some embodiments, the subject having BSCR has one copy of each HLA-A*30 and HLA-A*31. In some embodiments, the subject having BSCR has one copy of each HLA-A*30 and HLA-A*33. In some embodiments, the subject having BSCR has one copy of each HLA-A*31 and HLA-A*33.

In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*30. In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*31. In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*33. In some embodiments, the subject having BSCR has one copy of HLA-A*30 and two copies of HLA-A*31. In some embodiments, the subject having BSCR has one copy of HLA-A*30 and two copies HLA-A*33. In some embodiments, the subject having BSCR has one copy of HLA-A*31 and two copies of HLA-A*33.

In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*30. In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*31. In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*33. In some embodiments, the subject having BSCR has two copies of HLA-A*30 and one copy of HLA-A*31. In some embodiments, the subject having BSCR has two copies of HLA-A*30 and one copy of HLA-A*33. In some embodiments, the subject having BSCR has two copies of HLA-A*31 and one copy of HLA-A*33.

In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*30. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*31. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*33. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*30 and two copies of HLA-A*31. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA- A*30 and two copies of HLA-A*33. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*31 and two copies of HLA-A*33.

In some embodiments, the method further comprises administering to the subject an HLA-Awl9 inhibitor. In some embodiments, the HLA-Awl9 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-A*29 antibody. In some embodiments, the HLA- Awl9 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA (shRNA) that hybridizes to an HLA-Awl9. In some embodiments, the HLA-Awl9 is HLA-A*29.

In some embodiments, the MHC-l-opathy is AS. In some embodiments, the method further comprises detecting the presence or absence of HLA-B*27 or HLA-B*40 in a biological sample obtained from the subject. In some embodiments, the subject is or is suspected of being HLA-B*27 + . In some embodiments, the subject is or is suspected of being HLA-B*40 + . In some embodiments, the method further comprises determining whether the subject has one or two copies of HLA-B*27 or HLA-B*40. In some embodiments, the subject has a single copy of HLA-B*27 or HLA-B*40. In some embodiments, the subject has two copies of HLA-B*27 or HLA- B*40. In some embodiments, the method further comprises administering to the subject an HLA-B*27 inhibitor or an HLA-B*40 inhibitor. In some embodiments, the HLA-B*27 inhibitor or HLA-B*40 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*27 antibody or an anti-HLA-B*40 antibody. In some embodiments, the HLA-B*27 inhibitor or HLA- B*40 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*27 or an HLA-B*40.

In some embodiments, the MHC-l-opathy is Behget's disease. In some embodiments, the method further comprises detecting the presence or absence of HLA-B*51 in a biological sample obtained from the subject. In some embodiments, the subject is or is suspected of being HLA-B*51 + . In some embodiments, the method further comprises determining whether the subject has one or two copies of HLA-B*51. In some embodiments, the subject has a single copy of HLA-B*51. In some embodiments, the subject has two copies of HLA-B*51. In some embodiments, the method further comprises administering to the subject an HLA-B*51 inhibitor. In some embodiments, the HLA-B*51 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*51 antibody. In some embodiments, the HLA-B*51 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*51.

In some embodiments, the MHC-l-opathy is psoriasis. In some embodiments, the method further comprises detecting the presence or absence of HLA-C*06 in a biological sample obtained from the subject. In some embodiments, the subject is or is suspected of being HLA-C*06 + . In some embodiments, the method further comprises determining whether the subject has one or two copies of HLA-C*06. In some embodiments, the subject has a single copy of HLA-C*06. In some embodiments, the subject has two copies of HLA-C*06. In some embodiments, the method further comprises administering to the subject an HLA-C*06 inhibitor. In some embodiments, the HLA-C*06 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-C*06 antibody. In some embodiments, the HLA-C*06 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-C*06.

In some embodiments, the MHC-l-opathy is JIA. In some embodiments, the method further comprises detecting the presence or absence of HLA-B*27 and/or DRB1 in a biological sample obtained from the subject. In some embodiments, the subject is or is suspected of being HLA-B*27 + and/or DRB1 + . In some embodiments, the method further comprises determining whether the subject has one or two copies of HLA-B*27 and/or DRB1. In some embodiments, the subject has a single copy of HLA-B*27 and/or DRB1. In some embodiments, the subject has two copies of HLA-B*27 and/or DRB1. In some embodiments, the method further comprises administering to the subject an HLA-B*27 inhibitor and/or a DRB1 inhibitor. In some embodiments, the HLA-B*27 inhibitor and/or DRB1 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*27 antibody or an anti-DRBl antibody. In some embodiments, the HLA-B*27 inhibitor and/or DRB1 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*27 and/or an DRB1.

In some embodiments, the MHC-l-opathy is IBD or CD. In some embodiments, the method further comprises detecting the presence or absence of HLA-C*07 in a biological sample obtained from the subject. In some embodiments, the subject is or is suspected of being HLA-C*07 + . In some embodiments, the method further comprises determining whether the subject has one or two copies of HLA-C*07. In some embodiments, the subject has a single copy of HLA-C*07. In some embodiments, the subject has two copies of HLA-C*07. In some embodiments, the method further comprises administering to the subject an HLA-C*07 inhibitor. In some embodiments, the HLA-C*07 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-C*07 antibody. In some embodiments, the HLA-C*07 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-C*07.

In some embodiments, the ERAP2 inhibitor comprises a small molecule degrader, a proteoloysis-targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an siRNA that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an shRNA that hybridizes to ERAP2 mRNA. In some embodiments, the ERAP2 inhibitor comprises an anti-ERAP2 antibody. In some embodiments, the ERAP2 inhibitor comprises a pseudopeptide. In some embodiments, the pseudopeptide is a phosphinic pseudopeptide. In some embodiments, the phosphinic pseudopeptide is DG002 or DG013 (see, for example, Zervoudi et a I., Proc. Natl. Acad. Sci. USA, 2013, 110, 19890-19895). In some embodiments, the phosphinic pseudopeptide is DG002. In some embodiments, the phosphinic pseudopeptide is DG013. In some embodiments, the ERAP2 inhibitor comprises a small molecule. In some embodiments, the ERAP2 inhibitor is compound 4, compound 15, compound 16, compound 5, or analogues of compound 5, which are drug-like carboxylic acids and bioisosters screened for enhanced selectivity for ERAP2 over ERAP1 (see, Medve et al., European Journal of Medicinal Chemistry, 2021, 211, 113053). In some embodiments, the ERAP2 inhibitor is a phosphonic or phosphinic acid compound with higher affinity for ERAP2 than ERAP1 (see, Weglarz-Tomczak et al., Bioorg. Med. Chem. Lett., 2016, 26, 4122-4126). Additional ERAP2 inhibitors are described in, for example, Georgiadis et al., Curr. Medic. Chem., 2019, 26, 2715-2729.

In any of the embodiments described herein, the methods can further comprise administering to the subject an ERAP1 agonist or inhibitor, depending upon the MHC-l-opathy. For AS and psoriasis, an ERAP1 inhibitor can be administered. For the remaining MHC-I- opathies, an ERAP1 agonist can be administered.

In some embodiments, the ERAP1 agonist comprises an oligonucleotide. In some embodiments, the oligonucleotide is ODN1826. In some embodiments, the ERAP1 agonist comprises a peptide. In some embodiments, the ERAP1 agonist comprises a lipopeptide. In some embodiments, the lipopeptide is Pam3CSK4 or FSL-1. In some embodiments, the lipopeptide is Pam3CSK4. In some embodiments, the lipopeptide is FSL-1. In some embodiments, the ERAP1 agonist comprises a small molecule. In some embodiments, the ERAP1 agonist can comprise an ERAPl-specific transcriptional activator, an ERAP1 protein stabilizer, an agonist of ERAP1 enzymatic activity, or an activator of ERAP1 secretion. In some embodiments, the ERAP1 agonist can comprise an ERAPl-specific transcriptional activator. In some embodiments, the ERAP1 agonist can comprise an ERAP1 protein stabilizer. In some embodiments, the ERAP1 agonist can comprise an agonist of ERAP1 enzymatic activity. In some embodiments, the ERAP1 agonist can comprise an activator of ERAP1 secretion. Additional examples of ERAP1 agonists are described in, for example, Goto et al., J. Immunol., 2014, 192, 4443-4452.

In some embodiments, the ERAP1 inhibitor comprises a small molecule degrader, a proteoloysis-targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an siRNA that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an shRNA that hybridizes to ERAP1 mRNA. In some embodiments, the ERAP1 inhibitor comprises an anti-ERAPl antibody. In some embodiments, the ERAP1 inhibitor is DG002 and DG013 (see, Zervoudi et al., Proc. Nat'l Acad. Sci. USA, 2013, 110, 19890-19895). In some embodiments, the ERAP1 inhibitor is a phosphinic dipeptide or tripeptide analog (see, Weglarz-Tomczak et al., Bioorg. Med. Che, Lett., 2016, 26, 4122-4126). In some embodiments, the ERAP1 inhibitor is (N-(N-(2-(lH-indol-3-yl)ethyl)carbamimidoyl) -2,5-difluorobenzenesulfonamide), (l-(l-(4-acetylpiperazine-l-carbonyl)cyclohexyl) -3-(p-tolyl)urea), or (4-methoxy-3-(N-(2- (piperidin-l-yl)-5-(trifluoromethyl)phenyl) sulfamoyl)benzoic acid (see, Maben et al., J. Med. Chem., 2020, 63, 103-121). In some embodiments, the ERAP1 inhibitor is (4aR,5S,6R,8S,8aR)- 5-(2-(Furan-3-yl)ethyl)-8-hydroxy-5,6,8a-trimethyl-3,4,4a,5, 6,7,8,8a-octahydronaphthalene-l- carboxylic acid (see, Liddle et al., J. Med. Chem., 2020, 63, 3348-3358). In some embodiments, the ERAP1 inhibitor is DG013A or a phosphinic tripeptide or dipeptide or an aminophosphonic derivative, or 3,4-diaminobenzoic (DABA) derivative, or a derivative of thimerosal, (see, Georgiadis et al., Cur. Med. Chem., 2019, 26, 2715-2729). In some embodiments, the ERAP1 inhibitor is a benzofuran or 7-Benzofuran amide variation (see, Deddouche-Grass et al., ACS Med. Chem. Lett., 2021, 12, 1137-1142).

In any of the embodiments described herein, any of the inhibitors or other agents described herein can form a component of an antibody-drug-conjugate (ADC). For example, an ERAP1 inhibitor or an ERAP2 inhibitor can be conjugated to an antibody, or antigen-binding fragment thereof. The inhibitor can comprise a small molecule degrader, a proteoloysis- targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule.

The present disclosure also provides methods of treating a subject having an MHC-I- opathy. In some embodiments, the method comprises performing or having performed an assay on a biological sample from the subject to determine whether the subject comprises: i) an MHC-l-opathy-related HLA genotype; and ii) a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an ERAP2 inhibitor, wherein the subject comprises both an MHC-l-opathy-related HLA genotype and a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein. The presence of both the MHC-l-opathy-related HLA genotype and the functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein indicates that the subject is a candidate for treating the MHC-l-opathy by inhibiting ERAP2.

In some embodiments, the MHC-l-opathy is BSCR and the MHC-l-opathy-related HLA genotype comprises an HLA-Awl9 allele. In some embodiments, the HLA-Awl9 allele is an HLA- A*29 allele, an HLA-A*30 allele, an HLA-A*31 allele, or an HLA-A*33 allele, or any combination thereof. In some embodiments, the subject has a single copy of the HLA-Awl9 allele. In some embodiments, the HLA-Awl9 allele is an HLA-A*29 allele. In some embodiments, the HLA-Awl9 allele is an HLA-A*30 allele. In some embodiments, the HLA-Awl9 allele is an HLA-A*31 allele.

In some embodiments, the HLA-Awl9 allele is an HLA-A*33 allele. In some embodiments, the subject has two copies of the HLA-Awl9 allele. In some embodiments, the subject is or is suspected of being HLA-A*29 + /HLA-A*30 + . In some embodiments, the subject is or is suspected of being HLA-A*29 + /HLA-A*31 + . In some embodiments, the subject is or is suspected of being H LA-A* 29 + /H LA-A*33 + .

In some embodiments, the subject having BSCR is not HLA-A*29 + .

In some embodiments, the subject having BSCR has a copy of at least any two of HLA- A*29, HLA-A*30, HLA-A*31, or HLA-A*33. In some embodiments, the subject having BSCR has a copy of at least any three of HLA-A*29, HLA-A*30, HLA-A*31, or HLA-A*33. In some embodiments, the subject having BSCR has a copy of all of HLA-A*29, HLA-A*30, HLA-A*31, or HLA-A*33.

In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*30. In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*31. In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*33. In some embodiments, the subject having BSCR has one copy of each HLA-A*30 and HLA-A*31. In some embodiments, the subject having BSCR has one copy of each HLA-A*30 and HLA-A*33. In some embodiments, the subject having BSCR has one copy of each HLA-A*31 and HLA-A*33.

In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*30. In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*31. In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*33. In some embodiments, the subject having BSCR has one copy of HLA-A*30 and two copies of HLA-A*31. In some embodiments, the subject having BSCR has one copy of HLA-A*30 and two copies HLA-A*33. In some embodiments, the subject having BSCR has one copy of HLA-A*31 and two copies of HLA-A*33.

In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*30. In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*31. In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*33. In some embodiments, the subject having BSCR has two copies of HLA-A*30 and one copy of HLA-A*31. In some embodiments, the subject having BSCR has two copies of HLA-A*30 and one copy of HLA-A*33. In some embodiments, the subject having BSCR has two copies of HLA-A*31 and one copy of HLA-A*33.

In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*30. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*31. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*33. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*30 and two copies of HLA-A*31. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA- A*30 and two copies of HLA-A*33. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*31 and two copies of HLA-A*33.

In some embodiments, the method further comprises administering to the subject an HLA-Awl9 inhibitor. In some embodiments, the HLA-Awl9 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-A*29 antibody. In some embodiments, the HLA- Awl9 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-Awl9. In some embodiments, the HLA-Awl9 is H LA -A* 29.

In some embodiments, the MHC-l-opathy is AS and the MHC-l-opathy-related HLA genotype comprises an HLA-B*27 allele or an HLA-B*40 allele. In some embodiments, the subject has a single copy of HLA-B*27 or HLA-B*40. In some embodiments, the subject has two copies of HLA-B*27 or HLA-B*40. In some embodiments, the method further comprises administering to the subject an HLA-B*27 inhibitor or an HLA-B*40 inhibitor. In some embodiments, the HLA-B*27 inhibitor or HLA-B*40 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*27 antibody or an anti-HLA-B*40 antibody. In some embodiments, the HLA-B*27 inhibitor or HLA-B*40 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*27 or an HLA-B*40.

In some embodiments, the MHC-l-opathy is Behget's disease and the MHC-l-opathy- related HLA genotype comprises an HLA-B*51 allele. In some embodiments, the subject has a single copy of HLA-B*51. In some embodiments, the subject has two copies of HLA-B*51. In some embodiments, the method further comprises administering to the subject an HLA-B*51 inhibitor. In some embodiments, the HLA-B*51 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*51 antibody. In some embodiments, the HLA-B*51 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*51.

In some embodiments, the MHC-l-opathy is psoriasis and the MHC-l-opathy-related HLA genotype comprises an HLA-C*06 allele. In some embodiments, the subject has a single copy of HLA-C*06. In some embodiments, the subject has two copies of HLA-C*06. In some embodiments, the method further comprises administering to the subject an HLA-C*06 inhibitor. In some embodiments, the HLA-C*06 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-C*06 antibody. In some embodiments, the HLA-C*06 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-C*06.

In some embodiments, the MHC-l-opathy is JIA and the MHC-l-opathy-related HLA genotype comprises an HLA-B*27 and/or DRB1. In some embodiments, the subject has a single copy of HLA-B*27 and/or DRB1. In some embodiments, the subject has two copies of HLA-B*27 and/or. In some embodiments, the method further comprises administering to the subject an HLA-B*27 inhibitor and/or a DRB1 inhibitor. In some embodiments, the HLA-B*27 inhibitor and/or DRB1 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*27 antibody and/or a DRB1 antibody. In some embodiments, the HLA-B*27 inhibitor and/or DRB1 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*27 or DRB1.

In some embodiments, the MHC-l-opathy is IBD or CD and the MHC-l-opathy-related HLA genotype comprises an HLA-C*07 allele. In some embodiments, the subject has a single copy of HLA-C*07. In some embodiments, the subject has two copies of HLA-C*07. In some embodiments, the method further comprising administering to the subject an HLA-C*07 inhibitor. In some embodiments, the HLA-C*07 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-C*07 antibody. In some embodiments, the HLA-C*07 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-C*07.

In any of the embodiments described herein, the nucleic acid molecule comprises genomic DNA, mRNA, or cDNA obtained from mRNA. In some embodiments, the nucleic acid molecule comprises genomic DNA. In some embodiments, the nucleic acid molecule comprises mRNA. In some embodiments, the nucleic acid molecule comprises cDNA obtained from mRNA.

In any of the embodiments described herein, the ERAP2 inhibitor comprises a small molecule degrader, a proteoloysis-targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an siRNA that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an shRNA that hybridizes to ERAP2 mRNA. In some embodiments, the ERAP2 inhibitor comprises an anti-ERAP2 antibody. In some embodiments, the ERAP2 inhibitor comprises a pseudopeptide. In some embodiments, the pseudopeptide is a phosphinic pseudopeptide. In some embodiments, the phosphinic pseudopeptide is DG002 or DG013. In some embodiments, the ERAP2 inhibitor comprises a small molecule.

In any of the embodiments described herein, the assay performed or having been performed on the biological sample from the subject can further determine whether the subject comprises a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein. In some embodiments, the method further comprises administering to the subject a therapeutically effective amount of an ERAP1 agonist or inhibitor (depending upon the MHC-l-opathy), wherein the subject comprises an MHC-l-opathy-related HLA genotype and does or does not comprise a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein. The presence of an MHC-l-opathy-related HLA genotype and the absence of a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein indicates that the subject is a candidate for treating the MHC-l-opathy by activating ERAP1. The presence of an MHC-l-opathy-related HLA genotype and the presence of a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein indicates that the subject is a candidate for treating the MHC-l-opathy by inhibiting ERAP1. ln any of the embodiments described herein, the ERAP1 agonist comprises an oligonucleotide. In some embodiments, the oligonucleotide is ODN1826. In some embodiments, the ERAP1 agonist comprises a peptide. In some embodiments, the ERAP1 agonist comprises a lipopeptide. In some embodiments, the lipopeptide is Pam3CSK4 or FSL-1. In some embodiments, the lipopeptide is Pam3CSK4. In some embodiments, the lipopeptide is FSL-1. In some embodiments, the ERAP1 agonist comprises a small molecule. In some embodiments, the ERAP1 agonist can comprise an ERAPl-specific transcriptional activator, an ERAP1 protein stabilizer, an agonist of ERAP1 enzymatic activity, or an activator of ERAP1 secretion. In some embodiments, the ERAP1 agonist can comprise an ERAPl-specific transcriptional activator. In some embodiments, the ERAP1 agonist can comprise an ERAP1 protein stabilizer. In some embodiments, the ERAP1 agonist can comprise an agonist of ERAP1 enzymatic activity. In some embodiments, the ERAP1 agonist can comprise an activator of ERAP1 secretion. Additional examples of ERAP1 agonists are described in, for example, Goto et al., J. Immunol., 2014, 192, 4443-4452.

In any of the embodiments described herein, the ERAP1 inhibitor comprises a small molecule degrader, a proteoloysis-targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an siRNA that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an shRNA that hybridizes to ERAP1 mRNA. In some embodiments, the ERAP1 inhibitor comprises an anti-ERAPl antibody.

HLA-class-l antibodies can be generated by numerous methodologies with different degrees of antigen/allele specificity attained and are reported to be used for in vitro assays. HLA-B*27 antibodies can be generated by numerous methodologies. In addition, three commercially available antibodies for HLA-B27 flow cytometric screening include the monoclonal mouse anti-human ABC-m3, FD705, and GS145.2 which have been shown to each have differing levels of cross-reactivity to other HLA-B antigens/alleles (Levering et al., Cytometry B Clin. Cytom., 2003, 54, 28-38). HLA-B*51 antibodies can also be generated by numerous methodologies. For example, antibodies to a broader HLA-Bw4 epitope can be obtained from clone REA274 (e.g., HLA-B members: B5, B5102, B5103, B13, B17, B37, B38, B44, B47, B49, B51, B52, B53, B57, B58, B59, B63, and B77; HLA-A members: A9, A23, A24, A25, and A*32). In addition, antibodies to HLA-B*51/B*52/B*35 can be obtained from clone HDG8D9 (Drabbels et al., Blood, 2011, 118, el49-55). HLA-C*06 antibodies can also be generated by numerous methodologies. For example, pan HLA-C antibodies can be obtained from clone DT-9 (which also recognizes HLA-E) (Braud et al., Curr. Biol., 1998, 8, 1-10). Broad anti-HLA-C antibodies can be obtained from clone L31 (which also recognizes some HLA-B alleles) (Setini et al., Hum. Immunol., 1996, 46, 69-81). HLA-Cw6 scFv can also be generated which has weak binding to HLA-Cw2,4,5 (Marget et al., Mol. Immunol., 2005, 42, 643-649).

In some embodiments, the assay for determining whether the subject comprises an MHC-l-opathy-related and/or MHC-ll-opathy-related HLA genotype and a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein, is a genotyping assay or sequencing assay. In some embodiments, the nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein comprises genomic DNA, mRNA, or cDNA obtained from mRNA. By comparing the nucleotide or protein sequence of the ERAP2 protein and/or ERAP1 protein in the sample from a subject to the wild type sequence for ERAP2 protein and/or ERAP1 protein or nucleic acid molecule, or to published sequences of variant ERAP2 proteins and/or ERAP1 proteins or nucleic acid molecules having reduced or no activity, a determination can be made whether the subject comprises a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein. In addition, although an individual ERAP2 protein and/or ERAP1 protein may have biological activity, the overall function of the ERAP2 protein and/or ERAP1 protein may not be functional due to reduced levels of expression. Thus, as used herein, an ERAP2 protein and/or ERAP1 protein can be determined not to be functional because the ERAP2 protein and/or ERAP1 protein lacks or had reduced biological activity or because the expression level is reduced.

Determining whether a subject has an MHC-l-opathy-related and/or MHC-ll-opathy- related HLA genotype and/or a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein, in a biological sample from a subject can be carried out by any of the methods described herein. In some embodiments, these methods can be carried out in vitro. In some embodiments, these methods can be carried out in situ. In some embodiments, these methods can be carried out in vivo. In any of these embodiments, the nucleic acid molecule can be present within a biological sample obtained from the subject.

The biological sample can be derived from any cell, tissue, or biological fluid from the subject. The biological sample may comprise any clinically relevant tissue, such as a bone marrow sample, a tumor biopsy, a fine needle aspirate, or a sample of bodily fluid, such as blood, gingival crevicular fluid, plasma, serum, lymph, ascitic fluid, cystic fluid, or urine. In some cases, the sample comprises a buccal swab. The biological sample used in the methods disclosed herein can vary based on the assay format, nature of the detection method, and the tissues, cells, or extracts that are used as the sample. A biological sample can be processed differently depending on the assay being employed. For example, when detecting any particular nucleic acid molecule, preliminary processing designed to isolate or enrich the biological sample for the particular nucleic acid molecule can be employed. A variety of techniques may be used for this purpose. Various methods to detect the presence or level of an mRNA molecule or the presence of a particular genomic DNA locus can be used.

In some embodiments, the biological sample comprises a cell or cell lysate. Such methods can further comprise, for example, obtaining a biological sample from the subject comprising genomic nucleic acid molecules or mRNA molecules, and if mRNA, optionally reverse transcribing the mRNA into cDNA. In some embodiments, the method is an in vitro method. In some embodiments, the assay comprises RNA sequencing (RNA-Seq). In some embodiments, the assays also comprise reverse transcribing mRNA into cDNA, such as by the reverse transcriptase polymerase chain reaction (RT-PCR).

Illustrative examples of nucleic acid sequencing techniques include, but are not limited to, chain terminator (Sanger) sequencing and dye terminator sequencing. Other methods involve nucleic acid hybridization methods other than sequencing, including using labeled primers or probes directed against purified DNA, amplified DNA, and fixed cell preparations (fluorescence in situ hybridization (FISH)). In some methods, a target nucleic acid molecule may be amplified prior to or simultaneous with detection. Illustrative examples of nucleic acid amplification techniques include, but are not limited to, polymerase chain reaction (PCR), ligase chain reaction (LCR), strand displacement amplification (SDA), and nucleic acid sequence based amplification (NASBA). Other methods include, but are not limited to, ligase chain reaction, strand displacement amplification, and thermophilic SDA (tSDA). Administration of any of the therapeutic agents described herein (including the ERAP2 inhibitor, the ERAP1 agonist or inhibitor, and/or the HLA inhibitor) can be in a therapeutically effective amount to be determined by a health care professional. Administration of any of the therapeutic agents can be repeated, for example, after one day, two days, three days, five days, one week, two weeks, three weeks, one month, five weeks, six weeks, seven weeks, eight weeks, two months, or three months. The repeated administration can be at the same dose or at a different dose. The administration can be repeated once, twice, three times, four times, five times, six times, seven times, eight times, nine times, ten times, or more. For example, according to certain dosage regimens a subject can receive therapy for a prolonged period of time such as, for example, 6 months, 1 year, or more.

Administration of any of the therapeutic agents can occur by any suitable route including, but not limited to, parenteral, intravenous, oral, subcutaneous, intra-arterial, intracranial, intrathecal, intraperitoneal, topical, intranasal, intra-articular, intravitreal, intracameral, subretinal, suprachoroidal, or intramuscular.

Pharmaceutical compositions for administration are desirably sterile and substantially isotonic and manufactured under GMP conditions. Pharmaceutical compositions can be provided in unit dosage form (i.e., the dosage for a single administration). Pharmaceutical compositions can be formulated using one or more physiologically and pharmaceutically acceptable carriers, diluents, excipients or auxiliaries. The formulation depends on the route of administration chosen. The term "pharmaceutically acceptable" means that the carrier, diluent, excipient, or auxiliary is compatible with the other ingredients of the formulation and not substantially deleterious to the recipient thereof.

The terms "treat", "treating", and "treatment" and "prevent", "preventing", and "prevention" as used herein, refer to eliciting the desired biological response, such as a therapeutic and prophylactic effect, respectively. In some embodiments, a therapeutic effect comprises one or more of a decrease/reduction in an MHC-l-opathy and/or MHC-ll-opathy, a decrease/reduction in the severity of an MHC-l-opathy and/or MHC-ll-opathy (such as, for example, a reduction or inhibition of development of an MHC-l-opathy and/or MHC-ll-opathy), a decrease/reduction in symptoms and MHC-l-opathy-related effects and/or MHC-ll-opathy- related effects, delaying the onset of symptoms and MHC-l-opathy-related effects and/or MHC- ll-opathy-related effects, reducing the severity of symptoms of MHC-l-opathy-related effects and/or MHC-ll-opathy-related effects, reducing the severity of an acute episode, reducing the number of symptoms and MHC-l-opathy-related effects and/or MHC-ll-opathy-related effects, reducing the latency of symptoms and MHC-l-opathy-related effects and/or MHC-ll-opathy- related effects, an amelioration of symptoms and MHC-l-opathy-related effects and/or MHC-ll- opathy-related effects, reducing secondary symptoms, reducing secondary infections, preventing relapse to an MHC-l-opathy and/or MHC-ll-opathy, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, increasing time to sustained progression, speeding recovery, or increasing efficacy of or decreasing resistance to alternative therapeutics, and/or an increased survival time of the subject, following administration of the agent or composition comprising the agent. A prophylactic effect may comprise a complete or partial avoidance/inhibition or a delay of an MHC-l-opathy and/or MHC-ll-opathy development/progression (such as, for example, a complete or partial avoidance/inhibition or a delay), and an increased survival time of the affected subject, following administration of a therapeutic protocol. Treatment of an MHC-l-opathy and/or MHC- ll-opathy encompasses the treatment of subjects already diagnosed as having any form of the MHC-l-opathy and/or MHC-ll-opathy at any clinical stage or manifestation, the delay of the onset or evolution or aggravation or deterioration of the symptoms or signs of an MHC-l-opathy and/or MHC-ll-opathy, and/or preventing and/or reducing the severity of an MHC-l-opathy and/or MHC-ll-opathy.

In some embodiments, the antisense nucleic acid molecules targeted to ERAP2 comprise or consist of the nucleotide sequences shown in Table 1.

Table 1 DOCKET NO.: 189238.05102 (3358) (10814W001)

-23- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 24 -

In some embodiments, the antisense nucleic acid molecules targeted to ERAP2 comprise or consist of the nucleotide sequences shown in Table 2. Table 2 DOCKET NO.: 189238.05102 (3358) (10814W001)

-25- DOCKET NO.: 189238.05102 (3358) (10814W001)

-26- DOCKET NO.: 189238.05102 (3358) (10814W001)

-27- DOCKET NO.: 189238.05102 (3358) (10814W001)

-28- DOCKET NO.: 189238.05102 (3358) (10814W001)

-29- DOCKET NO.: 189238.05102 (3358) (10814W001)

-30- DOCKET NO.: 189238.05102 (3358) (10814W001)

-31- DOCKET NO.: 189238.05102 (3358) (10814W001)

-32- DOCKET NO.: 189238.05102 (3358) (10814W001)

-33- DOCKET NO.: 189238.05102 (3358) (10814W001)

-34- DOCKET NO.: 189238.05102 (3358) (10814W001)

-35- DOCKET NO.: 189238.05102 (3358) (10814W001)

-36- DOCKET NO.: 189238.05102 (3358) (10814W001)

-37- DOCKET NO.: 189238.05102 (3358) (10814W001)

-38- DOCKET NO.: 189238.05102 (3358) (10814W001)

-39- DOCKET NO.: 189238.05102 (3358) (10814W001)

-40- DOCKET NO.: 189238.05102 (3358) (10814W001)

-41- DOCKET NO.: 189238.05102 (3358) (10814W001)

-42- DOCKET NO.: 189238.05102 (3358) (10814W001)

-43- DOCKET NO.: 189238.05102 (3358) (10814W001)

-44- DOCKET NO.: 189238.05102 (3358) (10814W001)

-45- DOCKET NO.: 189238.05102 (3358) (10814W001)

-46- DOCKET NO.: 189238.05102 (3358) (10814W001)

-47- DOCKET NO.: 189238.05102 (3358) (10814W001)

-48- DOCKET NO.: 189238.05102 (3358) (10814W001)

-49- DOCKET NO.: 189238.05102 (3358) (10814W001)

-50- DOCKET NO.: 189238.05102 (3358) (10814W001)

-51- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 52 -

In some embodiments, the siRNA molecules targeted to ERAP2 comprise or consist of the nucleotide sequences (sense and antisense strands) shown in Table 3.

Table 3 DOCKET NO.: 189238.05102 (3358) (10814W001)

-53- DOCKET NO.: 189238.05102 (3358) (10814W001)

-54- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 55 -

In some embodiments, the siRNA molecules targeted to ERAP2 comprise or consist of the nucleotide sequences (sense and antisense strands) shown in Table 4.

Table 4 DOCKET NO.: 189238.05102 (3358) (10814W001)

-56- DOCKET NO.: 189238.05102 (3358) (10814W001)

-57- DOCKET NO.: 189238.05102 (3358) (10814W001)

-58- DOCKET NO.: 189238.05102 (3358) (10814W001)

-59- DOCKET NO.: 189238.05102 (3358) (10814W001)

-60- DOCKET NO.: 189238.05102 (3358) (10814W001)

-61- DOCKET NO.: 189238.05102 (3358) (10814W001)

-62- DOCKET NO.: 189238.05102 (3358) (10814W001)

-63- DOCKET NO.: 189238.05102 (3358) (10814W001)

-64- DOCKET NO.: 189238.05102 (3358) (10814W001)

-65- DOCKET NO.: 189238.05102 (3358) (10814W001) 66 DOCKET NO.: 189238.05102 (3358) (10814W001)

-67- DOCKET NO.: 189238.05102 (3358) (10814W001) 68 DOCKET NO.: 189238.05102 (3358) (10814W001)

-69- DOCKET NO.: 189238.05102 (3358) (10814W001)

-70- DOCKET NO.: 189238.05102 (3358) (10814W001)

-71- DOCKET NO.: 189238.05102 (3358) (10814W001)

-72- DOCKET NO.: 189238.05102 (3358) (10814W001)

-73- DOCKET NO.: 189238.05102 (3358) (10814W001)

-74- DOCKET NO.: 189238.05102 (3358) (10814W001)

-75- DOCKET NO.: 189238.05102 (3358) (10814W001)

-76- DOCKET NO.: 189238.05102 (3358) (10814W001)

-77- DOCKET NO.: 189238.05102 (3358) (10814W001)

-78- DOCKET NO.: 189238.05102 (3358) (10814W001)

-79- DOCKET NO.: 189238.05102 (3358) (10814W001)

-80- DOCKET NO.: 189238.05102 (3358) (10814W001)

-81- DOCKET NO.: 189238.05102 (3358) (10814W001)

-82- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 83 -

In some embodiments, the antisense nucleic acid molecules targeted to ERAP1 comprise or consist of the nucleotide sequences shown in Table 5.

Table 5 DOCKET NO.: 189238.05102 (3358) (10814W001)

-84- DOCKET NO.: 189238.05102 (3358) (10814W001)

-85- DOCKET NO.: 189238.05102 (3358) (10814W001) 86 DOCKET NO.: 189238.05102 (3358) (10814W001)

-87- DOCKET NO.: 189238.05102 (3358) (10814W001) 88 DOCKET NO.: 189238.05102 (3358) (10814W001)

-89- DOCKET NO.: 189238.05102 (3358) (10814W001)

-90- DOCKET NO.: 189238.05102 (3358) (10814W001)

-91- DOCKET NO.: 189238.05102 (3358) (10814W001)

-92- DOCKET NO.: 189238.05102 (3358) (10814W001)

-93- DOCKET NO.: 189238.05102 (3358) (10814W001)

-94- DOCKET NO.: 189238.05102 (3358) (10814W001)

-95- DOCKET NO.: 189238.05102 (3358) (10814W001)

-96- DOCKET NO.: 189238.05102 (3358) (10814W001)

-97- DOCKET NO.: 189238.05102 (3358) (10814W001)

-98- DOCKET NO.: 189238.05102 (3358) (10814W001)

-99- DOCKET NO.: 189238.05102 (3358) (10814W001) 100 DOCKET NO.: 189238.05102 (3358) (10814W001) 101 DOCKET NO.: 189238.05102 (3358) (10814W001) 102 DOCKET NO.: 189238.05102 (3358) (10814W001)

-103- DOCKET NO.: 189238.05102 (3358) (10814W001)

-104- DOCKET NO.: 189238.05102 (3358) (10814W001)

-105- DOCKET NO.: 189238.05102 (3358) (10814W001)

-106- DOCKET NO.: 189238.05102 (3358) (10814W001)

-107- DOCKET NO.: 189238.05102 (3358) (10814W001)

-108- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 109 -

In some embodiments, the siRNA molecules targeted to ERAP1 comprise or consist of the nucleotide sequences (sense and antisense strands) shown in Table 6.

Table 6 DOCKET NO.: 189238.05102 (3358) (10814W001) 110 DOCKET NO.: 189238.05102 (3358) (10814W001) 111 DOCKET NO.: 189238.05102 (3358) (10814W001) 112 DOCKET NO.: 189238.05102 (3358) (10814W001)

-113- DOCKET NO.: 189238.05102 (3358) (10814W001)

-114- DOCKET NO.: 189238.05102 (3358) (10814W001)

-115- DOCKET NO.: 189238.05102 (3358) (10814W001)

-116- DOCKET NO.: 189238.05102 (3358) (10814W001)

-117- DOCKET NO.: 189238.05102 (3358) (10814W001)

-118- DOCKET NO.: 189238.05102 (3358) (10814W001)

-119- DOCKET NO.: 189238.05102 (3358) (10814W001) 120 DOCKET NO.: 189238.05102 (3358) (10814W001) 121 DOCKET NO.: 189238.05102 (3358) (10814W001) 122 DOCKET NO.: 189238.05102 (3358) (10814W001)

-123- DOCKET NO.: 189238.05102 (3358) (10814W001)

-124- DOCKET NO.: 189238.05102 (3358) (10814W001)

-125- DOCKET NO.: 189238.05102 (3358) (10814W001)

-126- DOCKET NO.: 189238.05102 (3358) (10814W001)

-127- DOCKET NO.: 189238.05102 (3358) (10814W001)

-128- DOCKET NO.: 189238.05102 (3358) (10814W001)

-129- DOCKET NO.: 189238.05102 (3358) (10814W001)

-130- DOCKET NO.: 189238.05102 (3358) (10814W001)

-131- DOCKET NO.: 189238.05102 (3358) (10814W001)

-132- DOCKET NO.: 189238.05102 (3358) (10814W001)

-133- DOCKET NO.: 189238.05102 (3358) (10814W001)

-134- DOCKET NO.: 189238.05102 (3358) (10814W001)

-135- DOCKET NO.: 189238.05102 (3358) (10814W001)

-136- DOCKET NO.: 189238.05102 (3358) (10814W001)

-137- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 138 -

In some embodiments, the antisense nucleic acid molecules targeted to HLA-A comprise or consist of the nucleotide sequences shown in Table 7.

Table 7 DOCKET NO.: 189238.05102 (3358) (10814W001)

- 139 -

In some embodiments, the siRNA molecules targeted to HLA-A comprise or consist of the nucleotide sequences (sense and antisense strands) shown in Table 8.

Table 8 In some embodiments, the antisense nucleic acid molecules targeted to HLA-B comprise or consist of the nucleotide sequences shown in Table 9.

Table 9

In some embodiments, the siRNA molecules targeted to HLA-B comprise or consist of the nucleotide sequences (sense and antisense strands) shown in Table 10.

Table 10

In some embodiments, the antisense nucleic acid molecules targeted to HLA-C comprise or consist of the nucleotide sequences shown in Table 11. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 140 -

Table 11

In some embodiments, the siRNA molecules targeted to HLA-C comprise or consist of the nucleotide sequences (sense and antisense strands) shown in Table 12. Table 12

The inhibitory nucleic acid molecules disclosed herein can comprise RNA, DNA, or both RNA and DNA. The inhibitory nucleic acid molecules can also be linked or fused to a heterologous nucleic acid sequence, such as in a vector, or a heterologous label. For example, the inhibitory nucleic acid molecules disclosed herein can be within a vector or as an exogenous donor sequence comprising the inhibitory nucleic acid molecule and a heterologous nucleic acid sequence. The inhibitory nucleic acid molecules can also be linked or fused to a heterologous label. The label can be directly detectable (such as, for example, fluorophore) or indirectly detectable (such as, for example, hapten, enzyme, or fluorophore quencher). Such labels can be detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means. Such labels include, for example, radiolabels, pigments, dyes, chromogens, spin labels, and fluorescent labels. The label can also be, for example, a chemiluminescent substance; a metal-containing substance; or an enzyme, where there occurs an enzyme-dependent secondary generation of signal. The term "label" can also refer to a "tag" or hapten that can bind selectively to a conjugated molecule such that the conjugated molecule, when added subsequently along with a substrate, is used to generate a detectable signal. For example, biotin can be used as a tag along with an avidin or streptavidin conjugate of horseradish peroxidate ( H RP) to bind to the tag, and examined using a calorimetric substrate (such as, for example, tetramethylbenzidine (TMB)) or a DOCKET NO.: 189238.05102 (3358) (10814W001)

- 141 - fluorogenic substrate to detect the presence of HRP. Exemplary labels that can be used as tags to facilitate purification include, but are not limited to, myc, HA, FLAG or 3XFLAG, 6XHis or polyhistidine, glutathione-S-transferase (GST), maltose binding protein, an epitope tag, or the Fc portion of immunoglobulin. Numerous labels include, for example, particles, fluorophores, haptens, enzymes and their calorimetric, fluorogenic and chemiluminescent substrates and other labels.

The disclosed inhibitory nucleic acid molecules can comprise, for example, nucleotides or non-natural or modified nucleotides, such as nucleotide analogs or nucleotide substitutes. Such nucleotides include a nucleotide that contains a modified base, sugar, or phosphate group, or that incorporates a non-natural moiety in its structure. Examples of non-natural nucleotides include, but are not limited to, dideoxynucleotides, biotinylated, aminated, deaminated, alkylated, benzylated, and fluorophor-labeled nucleotides.

The inhibitory nucleic acid molecules disclosed herein can also comprise one or more nucleotide analogs or substitutions. A nucleotide analog is a nucleotide which contains a modification to either the base, sugar, or phosphate moieties. Modifications to the base moiety include, but are not limited to, natural and synthetic modifications of A, C, G, and T/U, as well as different purine or pyrimidine bases such as, for example, pseudouridine, uracil-5-yl, hypoxanthin-9-yl (I), and 2-aminoadenin-9-yl. Modified bases include, but are not limited to, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine,

6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo (such as, for example, 5-bromo), 5-trifluoromethyl and other 5-substituted uracils and cytosines,

7-methylguanine, 7-methyladenine, 8-azaguanine, 8-azaadenine, 7-deazaguanine, 7-deazaadenine, 3-deazaguanine, and 3-deazaadenine.

Nucleotide analogs can also include modifications of the sugar moiety. Modifications to the sugar moiety include, but are not limited to, natural modifications of DOCKET NO.: 189238.05102 (3358) (10814W001)

- 142 - the ribose and deoxy ribose as well as synthetic modifications. Sugar modifications include, but are not limited to, the following modifications at the 2' position: OH; F; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl, and alkynyl may be substituted or unsubstituted Cuoalkyl or C2-ioalkenyl, and C2-ioalkynyl. Exemplary 2' sugar modifications also include, but are not limited to, -0[(CH 2 )nO] m CH 3 , -0(CH 2 )nOCH 3 , -0(CH 2 )nNH 2 , -0(CH 2 )nCH 3 , -0(CH 2 )n-0NH 2 , and -0(CH 2 )nON[(CH 2 )nCH 3 )]2, where n and m, independently, are from 1 to about 10. Other modifications at the 2' position include, but are not limited to, Cuoalkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , S0 2 CH 3 , 0N0 2 , N0 2 , N 3 , NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. Similar modifications may also be made at other positions on the sugar, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide. Modified sugars can also include those that contain modifications at the bridging ring oxygen, such as CH2 and S. Nucleotide sugar analogs can also have sugar mimetics, such as cyclobutyl moieties in place of the pentofuranosyl sugar.

Nucleotide analogs can also be modified at the phosphate moiety. Modified phosphate moieties include, but are not limited to, those that can be modified so that the linkage between two nucleotides contains a phosphorothioate, chiral phosphorothioate, phosphorodithioate, phosphotriester, aminoalkylphosphotriester, methyl and other alkyl phosphonates including 3'-alkylene phosphonate and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates. These phosphate or modified phosphate linkage between two nucleotides can be through a 3'-5' linkage or a 2'-5' linkage, and the linkage can contain inverted polarity such as 3'-5' to 5'-3' or 2'-5' to 5'-2'. Various salts, mixed salts, and free DOCKET NO.: 189238.05102 (3358) (10814W001)

- 143 - acid forms are also included. Nucleotide substitutes also include peptide nucleic acids (PNAs).

In some embodiments, the antisense nucleic acid molecules are gapmers, whereby the first one to seven nucleotides at the 5' and 3' ends each have 2'-methoxyethyl (2'-MOE) modifications. In some embodiments, the first five nucleotides at the 5' and 3' ends each have 2'-MOE modifications. In some embodiments, the first one to seven nucleotides at the 5' and 3' ends are RNA nucleotides. In some embodiments, the first five nucleotides at the 5' and 3' ends are RNA nucleotides. In some embodiments, each of the backbone linkages between the nucleotides is a phosphorothioate linkage.

In some embodiments, the siRNA molecules have termini modifications. In some embodiments, the 5' end of the antisense strand is phosphorylated. In some embodiments, 5'-phosphate analogs that cannot be hydrolyzed, such as 5'-(E)-vinyl-phosphonate are used.

In some embodiments, the siRNA molecules have backbone modifications. In some embodiments, the modified phosphodiester groups that link consecutive ribose nucleosides have been shown to enhance the stability and in vivo bioavailability of siRNAs The non-ester groups (-OH, =0) of the phosphodiester linkage can be replaced with sulfur, boron, or acetate to give phosphorothioate, boranophosphate, and phosphonoacetate linkages. In addition, substituting the phosphodiester group with a phosphotriester can facilitate cellular uptake of siRNAs and retention on serum components by eliminating their negative charge. In some embodiments, the siRNA molecules have sugar modifications. In some embodiments, the sugars are deprotonated (reaction catalyzed by exo- and endonucleases) whereby the 2'-hydroxyl can act as a nucleophile and attack the adjacent phosphorous in the phosphodiester bond. Such alternatives include 2'-0-methyl, 2'-0-methoxyethyl, and 2'- fluoro modifications.

In some embodiments, the siRNA molecules have base modifications. In some embodiments, the bases can be substituted with modified bases such as pseudouridine, 5'- methylcytidine, N6-methyladenosine, inosine, and N7-methylguanosine.

In some embodiments, the siRNA molecules are conjugated to lipids. Lipids can be conjugated to the 5' or 3' termini of siRNA to improve their in vivo bioavailability by allowing DOCKET NO.: 189238.05102 (3358) (10814W001)

- 144 - them to associate with serum lipoproteins. Representative lipids include, but are not limited to, cholesterol and vitamin E, and fatty acids, such as palmitate and tocopherol.

In some embodiments, a representative siRNA has the following formula:

Sense: mN*mN*/i2FN/mN/i2FN/mN/i2FN/mN/i2FN/mN/i2FN/mN/i2FN/mN/i2FN/ mN/ i2FN/*mN*/32FN/

Antisense: /52FN/*/i2FN/*mN/i2FN/mN/i2FN/mN/i2FN/mN/i2FN/mN/i2FN/mN/i2F N/mN/ i2FN/mN/i2FN/mN*N*N wherein: "N" is the base; "2F" is a 2'-F modification; "m" is a 2'-0-methyl modification, "I" is an internal base; and "*" is a phosphorothioate backbone linkage.

The present disclosure also provides vectors comprising any one or more of the inhibitory nucleic acid molecules disclosed herein. In some embodiments, the vectors comprise any one or more of the inhibitory nucleic acid molecules disclosed herein and a heterologous nucleic acid. The vectors can be viral or nonviral vectors capable of transporting a nucleic acid molecule. In some embodiments, the vector is a plasmid or cosmid (such as, for example, a circular double-stranded DNA into which additional DNA segments can be ligated). In some embodiments, the vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Expression vectors include, but are not limited to, plasmids, cosmids, retroviruses, adenoviruses, adeno-associated viruses (AAV), plant viruses such as cauliflower mosaic virus and tobacco mosaic virus, yeast artificial chromosomes (YACs), Epstein-Barr (EBV)-derived episomes, and other expression vectors known in the art.

The present disclosure also provides compositions comprising any one or more of the inhibitory nucleic acid molecules disclosed herein. In some embodiments, the composition is a pharmaceutical composition. In some embodiments, the compositions comprise a carrier and/or excipient. Examples of carriers include, but are not limited to, poly(lactic acid) (PLA) microspheres, poly(D,L-lactic-coglycolic-acid) (PLGA) microspheres, liposomes, micelles, inverse micelles, lipid cochleates, and lipid microtubules. A carrier may comprise a buffered salt solution such as PBS, HBSS, etc.

The present disclosure also provides methods of identifying a subject having an increased risk for developing an MHC-l-opathy and/or an MHC-ll-opathy. The methods DOCKET NO.: 189238.05102 (3358) (10814W001)

- 145 - comprise performing or having performed an assay on a biological sample from the subject to determine whether the subject comprises: i) an MHC-l-opathy-related HLA genotype and/or an MHC-ll-opathy-related HLA genotype; and ii) a functional ERAP2 protein or a nucleic acid molecule encoding a functional ERAP2 protein. When the subject has both the MHC-l-opathy-related HLA genotype and/or an MHC-ll-opathy-related HLA genotype and the functional ERAP2 protein or the nucleic acid molecule encoding the functional ERAP2 protein, then the subject has an increased risk of developing the MHC-l-opathy and/or an MHC-ll-opathy. When the subject lacks the MHC-l-opathy-related HLA genotype and/or an MHC-ll-opathy-related HLA genotype, or lacks the functional ERAP2 protein or the nucleic acid molecule encoding the functional ERAP2 protein, or lacks both, then the subject has a decreased risk of developing the MHC-l-opathy and/or an MHC-ll-opathy. In some embodiments, the method further comprises determining whether the subject has a single copy of the MHC-l-opathy-related HLA genotype and/or an MHC-ll-opathy-related HLA genotype or two copies of the MHC-l-opathy-related HLA genotype and/or an MHC-ll- opathy-related HLA genotype. When the subject comprises two copies of the MHC-l-opathy- related HLA genotype and/or an MHC-ll-opathy-related HLA genotype, then the subject has an increased risk of developing the MHC-l-opathy and/or an MHC-ll-opathy compared to comprising a single copy of the MHC-l-opathy-related HLA genotype and/or an MHC-ll- opathy-related HLA genotype.

In some embodiments, the MHC-l-opathy is BSCR. In some embodiments, the subject is HLA-Awl9 + . In some embodiments, the subject is or is suspected of being HLA- A*29 + , HLA-A*30 + , HLA-A*31 + , or HLA-A*33 + , or any combination thereof. In some embodiments, the subject has a single copy of HLA-Awl9. In some embodiments, the subject has two copies of HLA-Awl9. In some embodiments, the subject is HLA-A*29 + /HLA- A*30 + . In some embodiments, the subject is HLA-A*29 + /HLA-A*31 + . In some embodiments, the subject is HLA-A*29 + /HLA-A*33 + .

In some embodiments, the subject having BSCR is not HLA-A*29 + .

In some embodiments, the subject having BSCR has a copy of at least any two of HLA-A*29, HLA-A*30, HLA-A*31, or HLA-A*33. In some embodiments, the subject having BSCR has a copy of at least any three of HLA-A*29, HLA-A*30, HLA-A*31, or HLA-A*33. In DOCKET NO.: 189238.05102 (3358) (10814W001)

- 146 - some embodiments, the subject having BSCR has a copy of all of HLA-A*29, HLA-A*30, HLA- A*31, or HLA-A*33.

In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*30. In some embodiments, the subject having BSCR has one copy of each HLA- A*29 and HLA-A*31. In some embodiments, the subject having BSCR has one copy of each HLA-A*29 and HLA-A*33. In some embodiments, the subject having BSCR has one copy of each HLA-A*30 and HLA-A*31. In some embodiments, the subject having BSCR has one copy of each HLA-A*30 and HLA-A*33. In some embodiments, the subject having BSCR has one copy of each HLA-A*31 and HLA-A*33.

In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*30. In some embodiments, the subject having BSCR has one copy of HLA- A*29 and two copies of HLA-A*31. In some embodiments, the subject having BSCR has one copy of HLA-A*29 and two copies of HLA-A*33. In some embodiments, the subject having BSCR has one copy of HLA-A*30 and two copies of HLA-A*31. In some embodiments, the subject having BSCR has one copy of HLA-A*30 and two copies HLA-A*33. In some embodiments, the subject having BSCR has one copy of HLA-A*31 and two copies of HLA- A*33.

In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*30. In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*31. In some embodiments, the subject having BSCR has two copies of HLA-A*29 and one copy of HLA-A*33. In some embodiments, the subject having BSCR has two copies of HLA-A*30 and one copy of HLA-A*31. In some embodiments, the subject having BSCR has two copies of HLA-A*30 and one copy of HLA-A*33. In some embodiments, the subject having BSCR has two copies of HLA-A*31 and one copy of HLA- A*33.

In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*30. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*31. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*29 and two copies of HLA-A*33. In some embodiments, the subject DOCKET NO.: 189238.05102 (3358) (10814W001)

- 147 - having BSCR or suspected of having BSCR has two copies of HLA-A*30 and two copies of HLA-A*31. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*30 and two copies of HLA-A*33. In some embodiments, the subject having BSCR or suspected of having BSCR has two copies of HLA-A*31 and two copies of HLA-A*33.

In some embodiments, the method further comprises administering to the subject an HLA-Awl9 inhibitor. In some embodiments, the HLA-Awl9 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-A*29 antibody. In some embodiments, the HLA-Awl9 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, a small interfering RNA (siRNA), or a short hairpin RNA (shRNA) that hybridizes to an HLA-Awl9. In some embodiments, the HLA-Awl9 is HLA-A*29.

In some embodiments, the MHC-l-opathy is AS. In some embodiments, the subject is or is suspected of being HLA-B*27 + or HLA-B*40 + . In some embodiments, the subject has a single copy of HLA-B*27 or HLA-B*40. In some embodiments, the subject has two copies of HLA-B*27 or HLA-B*40. In some embodiments, the method further comprises administering to the subject an HLA-B*27 inhibitor or an HLA-B*40 inhibitor. In some embodiments, the HLA-B*27 inhibitor or HLA-B*40 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*27 antibody or an anti-HLA-B*40 antibody. In some embodiments, the HLA-B*27 inhibitor or HLA-B*40 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*27 or HLA-B*40.

In some embodiments, the MHC-l-opathy is Behget's disease. In some embodiments, the subject is or is suspected of being HLA-B*51 + . In some embodiments, the subject has a single copy of HLA-B*51. In some embodiments, the subject has two copies of HLA-B*51. In some embodiments, the method further comprises administering to the subject an HLA-B*51 inhibitor. In some embodiments, the HLA-B*51 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*51 antibody. In some embodiments, the HLA-B*51 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid DOCKET NO.: 189238.05102 (3358) (10814W001)

- 148 - molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*51.

In some embodiments, the MHC-l-opathy is psoriasis. In some embodiments, the subject is or is suspected of being HLA-C*06 + . In some embodiments, the subject has a single copy of HLA-C*06. In some embodiments, the subject has two copies of HLA-C*06. In some embodiments, the method further comprises administering to the subject an HLA- C*06 inhibitor. In some embodiments, the HLA-C*06 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-C*06 antibody. In some embodiments, the HLA- C*06 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule.

In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-C*06.

In some embodiments, the MHC-l-opathy is JIA. In some embodiments, the subject is or is suspected of being HLA-B*27 + and/or DRB1 + . In some embodiments, the subject has a single copy of HLA-B*27 and/or DRB1. In some embodiments, the subject has two copies of HLA-B*27 and/or DRB1. In some embodiments, the method further comprises administering to the subject an HLA-B*27 inhibitor and/or a DRB1 inhibitor. In some embodiments, the HLA-B*27 inhibitor and/or DRB1 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-B*27 antibody or an anti-DRBl antibody. In some embodiments, the HLA-B*27 inhibitor and/or DRB1 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-B*27 and/or an DRB1.

In some embodiments, the MHC-l-opathy is IBD or CD. In some embodiments, the subject is or is suspected of being HLA-C*07 + . In some embodiments, the subject has a single copy of HLA-C*07. In some embodiments, the subject has two copies of HLA-C*07. In some embodiments, the method further comprises administering to the subject an HLA- C*07 inhibitor. In some embodiments, the HLA-C*07 inhibitor is an antibody. In some embodiments, the antibody is an anti-HLA-C*07 antibody. In some embodiments, the HLA- C*07 inhibitor comprises a small molecule degrader or an inhibitory nucleic acid molecule. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 149 -

In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to an HLA-C*07.

In any of the embodiments described herein, the methods can further comprise administering to the subject having an increased risk of developing the MHC-l-opathy- related HLA genotype and/or an MHC-ll-opathy an ERAP2 inhibitor. In some embodiments, the ERAP2 inhibitor comprises a small molecule degrader, a proteoloysis-targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an siRNA that hybridizes to ERAP2 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an shRNA that hybridizes to ERAP2 mRNA. In some embodiments, the ERAP2 inhibitor comprises an anti- ERAP2 antibody. In some embodiments, the ERAP2 inhibitor comprises a pseudopeptide. In some embodiments, the pseudopeptide is a phosphinic pseudopeptide. In some embodiments, the phosphinic pseudopeptide is DG002 or DG013. In some embodiments, the phosphinic pseudopeptide is DG002. In some embodiments, the phosphinic pseudopeptide is DG013. In some embodiments, the ERAP2 inhibitor comprises a small molecule.

In any of the embodiments described herein, the assay performed or having been performed on the biological sample from the subject can further determine whether the subject comprises a functional ERAP1 protein or a nucleic acid molecule encoding a functional ERAP1 protein. When the subject has the MHC-l-opathy-related HLA genotype and/or MHC-ll-opathy-related HLA genotype and lacks the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, then the subject has an increased risk of developing the MHC-l-opathy (for MHC-l-opathies except AS and psoriasis) and/or MHC-ll-opathy. When the subject has the MHC-l-opathy-related HLA genotype and/or MHC-ll-opathy-related HLA genotype and has a functional ERAP1 protein or a nucleic acid molecule encoding the functional ERAP1 protein, then the subject has an increased risk of developing the MHC-l-opathy (for AS and psoriasis). When the subject lacks the MHC-I- DOCKET NO.: 189238.05102 (3358) (10814W001)

- 150 - opathy-related HLA genotype and/or MHC-ll-opathy-related HLA genotype, or has the functional ERAP1 protein or the nucleic acid molecule encoding the functional ERAP1 protein, or both, then the subject has a decreased risk of developing the MHC-l-opathy and/or MHC-ll-opathy.

In any of the embodiments described herein, the methods can further comprise administering to the subject an ERAP1 agonist or inhibitor, depending upon the MHC-l- opathy. For AS and psoriasis, an ERAP1 inhibitor can be administered. For the remaining MHC-l-opathies, an ERAP1 agonist can be administered.

In some embodiments, the ERAP1 agonist comprises an oligonucleotide. In some embodiments, the oligonucleotide is ODN1826. In some embodiments, the ERAP1 agonist comprises a peptide. In some embodiments, the ERAP1 agonist comprises a lipopeptide. In some embodiments, the lipopeptide is Pam3CSK4 or FSL-1. In some embodiments, the lipopeptide is Pam3CSK4. In some embodiments, the lipopeptide is FSL-1. In some embodiments, the ERAP1 agonist comprises a small molecule. In some embodiments, the ERAP1 agonist can comprise an ERAPl-specific transcriptional activator, an ERAP1 protein stabilizer, an agonist of ERAP1 enzymatic activity, or an activator of ERAP1 secretion. In some embodiments, the ERAP1 agonist can comprise an ERAPl-specific transcriptional activator. In some embodiments, the ERAP1 agonist can comprise an ERAP1 protein stabilizer. In some embodiments, the ERAP1 agonist can comprise an agonist of ERAP1 enzymatic activity. In some embodiments, the ERAP1 agonist can comprise an activator of ERAP1 secretion. Additional examples of ERAP1 agonists are described in, for example, Goto et al., J. Immunol., 2014, 192, 4443-4452.

In some embodiments, the ERAP1 inhibitor comprises a small molecule degrader, a proteoloysis-targeting chimera, an immunomodulatory drug, or an inhibitory nucleic acid molecule. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule, an siRNA, or an shRNA that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an antisense nucleic acid molecule that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid molecule is an siRNA that hybridizes to ERAP1 mRNA. In some embodiments, the inhibitory nucleic acid DOCKET NO.: 189238.05102 (3358) (10814W001)

- 151 - molecule is an shRNA that hybridizes to ERAP1 mRNA. In some embodiments, the ERAP1 inhibitor comprises an anti-ERAPl antibody.

In some embodiments, the assay for determining whether the subject comprises an MHC-l-opathy-related and/or MHC-ll-opathy-related HLA genotype and a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein, is a genotyping assay or sequencing assay. In some embodiments, the nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein comprises genomic DNA, mRNA, or cDNA obtained from mRNA. By comparing the nucleotide or protein sequence of the ERAP2 protein and/or ERAP1 protein in the sample from a subject to the wild type sequence for ERAP2 protein and/or ERAP1 protein or nucleic acid molecule, or to published sequences of variant ERAP2 proteins and/or ERAP1 proteins or nucleic acid molecules having reduced or no activity, a determination can be made whether the subject comprises a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein. In addition, although an individual ERAP2 protein and/or ERAP1 protein may have biological activity, the overall function of the ERAP2 protein and/or ERAP1 protein may not be functional due to reduced levels of expression. Thus, as used herein, an ERAP2 protein and/or ERAP1 protein can be determined not to be functional because the ERAP2 protein and/or ERAP1 protein lacks or had reduced biological activity or because the expression level is reduced.

Determining whether a subject has an MHC-l-opathy-related and/or MHC-ll- opathy-related HLA genotype and/or a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein, in a biological sample from a subject can be carried out by any of the methods described herein. In some embodiments, these methods can be carried out in vitro. In some embodiments, these methods can be carried out in situ. In some embodiments, these methods can be carried out in vivo. In any of these embodiments, the nucleic acid molecule can be present within a biological sample obtained from the subject.

The biological sample can be derived from any cell, tissue, or biological fluid from the subject. The biological sample may comprise any clinically relevant tissue, such as a DOCKET NO.: 189238.05102 (3358) (10814W001)

- 152 - bone marrow sample, a tumor biopsy, a fine needle aspirate, or a sample of bodily fluid, such as blood, gingival crevicular fluid, plasma, serum, lymph, ascitic fluid, cystic fluid, or urine. In some cases, the sample comprises a buccal swab. The biological sample used in the methods disclosed herein can vary based on the assay format, nature of the detection method, and the tissues, cells, or extracts that are used as the sample. A biological sample can be processed differently depending on the assay being employed. For example, when detecting any particular nucleic acid molecule, preliminary processing designed to isolate or enrich the biological sample for the particular nucleic acid molecule can be employed. A variety of techniques may be used for this purpose. Various methods to detect the presence or level of an mRNA molecule or the presence of a particular genomic DNA locus can be used.

In some embodiments, the biological sample comprises a cell or cell lysate. Such methods can further comprise, for example, obtaining a biological sample from the subject comprising genomic nucleic acid molecules or mRNA molecules, and if mRNA, optionally reverse transcribing the mRNA into cDNA. In some embodiments, the method is an in vitro method. In some embodiments, the assay comprises RNA sequencing (RNA-Seq). In some embodiments, the assays also comprise reverse transcribing mRNA into cDNA, such as by the reverse transcriptase polymerase chain reaction (RT-PCR).

Detecting the presence or absence of any particular HLA allele can be carried out by numerous techniques. Detection of HLA-A alleles on a 2-digit and 4-digit resolution can be carried out. For example, an assay that targets the FI LA region in high resolution (all class-1 and class-ll genes) can be used. In some embodiments, the assay amplifies the full HLA gene (in this case HLA-A) from the 5'UTR to the 3'UTR and provides genetic variants across the full amplicon (the DNA that is the product of this amplification of the gene). A method can then be used to call the HLA-A alleles with high accuracy (e.g., PHLAT2; Bai et al., Methods Mol. Biol., 2018, 1802, 193-201). The output of PHLAT2 provides the HLA-A 4- digits allele data for each sample, which can be used for the analysis that identified other Awl9 alleles as enriched in Birdshot cases. In addition, commercial sources of HLA typing are available. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 153 -

Detecting the presence or absence a functional ERAP2 protein and/or ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP2 protein and/or ERAP1 protein, can be carried out by numerous techniques. For example, detection of presence or absence of ERAP2 protein and the relevant nucleotide sequence can be carried out as described in Andres et a I., PLoS Genetics, 2010, 6, 1-13. For example, a subject having an ERAP2 intronic variant designated rs2248374-A has a functional ERAP2 protein, or a nucleic acid molecule encoding a functional ERAP2 protein, and has an increased risk of developing the MHC-l-opathy. A subject having an ERAP2 variant designated rsl0044354, HapA has a functional ERAP2 protein, or a nucleic acid molecule encoding a functional ERAP2 protein, and has an increased risk of developing the MHC-l-opathy. In addition, a subject having an ERAP1 intronic variant designated rs27432-G does not have a functional ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP1 protein, and has an increased risk of developing the MHC-l-opathy. A subject having an ERAP1 variant designated rs2287987,

Hap 10 does not have a functional ERAP1 protein, or a nucleic acid molecule encoding a functional ERAP1 protein, and has an increased risk of developing the MHC-l-opathy. A subject having an ERAP2 splice variant designated rs2248374-G does not have a functional ERAP2 protein, or a nucleic acid molecule encoding a functional ERAP2 protein, and has a decreased risk of developing the MHC-l-opathy.

In order that the subject matter disclosed herein may be more efficiently understood, examples are provided below. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the claimed subject matter in any manner. Throughout these examples, molecular cloning reactions, and other standard recombinant DNA techniques, were carried out according to methods described in Maniatis et al., Molecular Cloning - A Laboratory Manual, 2nd ed., Cold Spring Harbor Press (1989), using commercially available reagents, except where otherwise noted.

Examples

Example 1: Methods

Study subjects and samples DOCKET NO.: 189238.05102 (3358) (10814W001)

- 154 -

The genomic DNA samples from 286 patients with BSCR and 108 unrelated healthy local French volunteers that exhibited HLA tissue typing common in the French population were included in this study. The patients were recruited at Hopital Cochin, Paris, France. All patients met the criteria for diagnosis of BSCR as defined both by an international consensus conference held in 2002 and by the Standardization of Uveitis Nomenclature (SUN) Working Group. In brief, all patients had a posterior bilateral uveitis with multifocal cream-colored or yellow-orange, oval or round choroidal lesions ("birdshot spots"). Although the presence of the FILA-A*29 allele was not a requirement for the diagnosis of BSCR according to the international criteria, all patients included in the current study carried the FILA-A*29 allele. The control DNA samples were collected from volunteer donors recruited by the hematopoietic stem cell donor center of Rheims for France Greffe de Moelle Registry, and local control healthy individuals of the Registry. The DNA samples were isolated from peripheral blood samples using a standard salting out method or QIAamp Blood Kit (Qiagen, Chatsworth, CA, USA). Quality and quantity of DNA was determined by UV spectrophotometry and the concentration was adjusted to 100 ng/ml. Signed informed consent documentation was obtained from all participants, and all research adhered to the tenets set forth in the Declaration of Helsinki. All study-related data acquisitions were approved by the Paris Cochin institutional review board.

Genetic data

A comprehensive approach was taken to both sequence the exomes and genotype all samples, to allow for identification of common and rare variants filtered based on high quality calls. DNA from participants was genotyped on the lllumina Global Screening Array (GSA) and imputed to the H RC reference panel. Prior to imputation, the variants that had a MAF >= 0.1%, missingness < 1% and FIWE p-value > 10 15 were retained. Imputation using the H RC reference panel yielded 8,385,561 variants with imputation INFO>0.3 and MAF>0.5%.

Exome sequencing was performed to a mean depth of 31X, followed by variant calling and quality control as reported previously (Van Flout et a I., Nature, 2020, 586, 749- 756), resulting in 238,942 variants. When integrated, this produced an overall dataset with DOCKET NO.: 189238.05102 (3358) (10814W001)

- 155 -

8,459,907 variants: 65.5% common (MAF > 5%), 34.5% low-frequency (0.5% < MAF < 5%) and 0.01% rare (MAF < 0.5%).

HLA genotyping

FI LA Class I genes (HLA-A, -B, and -C) were amplified in a multiplex PCR reaction with primers encompassing the full genomic loci for each target. The resulting amplicons were enzymatically fragmented to an average size of 250 base pairs and prepared for lllumina sequencing (New England Biolabs, Ipswich, MA). The libraries were sequenced on the lllumina HiSeq 2500 platform on a rapid run flow cell using paired-end 125 base pair reads with dual 10 base pair indexes. Upon completion of sequencing, raw data from each lllumina HiSeq run was gathered in local buffer storage and uploaded to the DNAnexus platform (Reid et a I., BMC Bioinformatics, 2014, 15, 30) for automated analysis. The FASTQ- formatted reads were converted from the BCL files and assigned to samples identified by specific barcodes using the bcl2fastq conversion software (lllumina Inc., San Diego, CA). All the reads in sample-specific FASTQ files were subject to HLA typing analysis using an updated version of PHLAT program (Bai et al., BMC Genomics, 2014, 15, 325) with the reference sequences consisting of GRCh38 genomic sequences and HLA type reference sequences in the IPD-IMGT/HLA database v3.30.0 (Robinson et al., Hum. Immunol., 2016, 77, 233-237).

In addition, HLA allele imputation was performed following SNP2HLA (Jia et al., PLoS One, 2013, 8, e64683) with the T1DGC HLA allele reference panel (Rich et al., Ann. N.Y. Acad. Sci., 2006, 1079, 1-8). HRC-imputed genotypes in the extended Major Histocompatibility Complex (MHC) region (chr6:25-35Mb) were filtered for high INFO score (>0.9) and certainty (maximum GP>0.8 for all genotyped), in order to increase overlap with the T1DGC reference panel, were re-phased along with chromosome 6 array genotypes using SHAPEIT4 (Delaneau et al., Nat. Commun., 2019, 10, 5436), and were imputed using Minimac4 (Das et al., Nat. Genet., 2016, 48, 1284-1287). HLA allele imputation quality was assessed by examining INFO score vs MAF, and imputed vs reference panel MAF.

Genetic association analyses

Association analyses in each study were performed using the genome-wide Firth logistic regression test implemented in SAIGE (Mbatchou et al., bioRxiv, 2020, DOCKET NO.: 189238.05102 (3358) (10814W001)

- 156 -

2020.2006.2019.162354, doi:10.1101/2020.06.19.162354; and Zhou et al., Nat. Genet.,

2018, 50, 1335-1341). In this implementation, Firth's approach is applied when the p-value from standard logistic regression score test is below 0.05. The directly genotyped variants with a minor allele frequency (MAF) >1%, <10% missingness, Flardy-Weinberg equilibrium test P-value>10 15 and linkage-disequilibrium (LD) pruning (1000 variant windows, 100 variant sliding windows and r 2 <0.1) were included for GRM for SAIGE. The association model included as covariates sex and the first 10 ancestry-informative principal components (PCs) derived from the GRM dataset. Flaplotype analyses were performed using PLINK 1.0 (Purcell et al., Am. J. Flum. Genet., 2007, 81, 559-575) -chap and -hap-assoc and -hap-logistic, and in R. High haplotype imputation and phasing quality was indicated by PLINK -hap-phase maximum likelihood haplotype genotypes' posterior probabilities all equal to one.

HLA-A allele association analyses

Association of HLA-A alleles was performed as follows: for each sample, both HLA-A alleles were typed as described above. Following HLA allele typing, related samples were removed. For the remaining cohort of 282 cases and 106 controls, one HLA-A allele that is not A*29 (the "second" allele) was obtained next. Samples carrying two copies of A*29, were considered having A*29 as the second allele. The cohort was then subjected to a Fisher's exact test, which tested the association of each allele that was identified in three or more BSCR cases, with the case-control status. To answer the question of whether the A19 allele group is also associated with the case-control status, the samples were combined, and tested together in two different ways: carrying all Awl9 alleles (A*29, A*30, A*31, A*32 and A*33). Since A*32 is biologically different than the other Awl9 alleles in its peptide binding domain, a group that is made of samples carrying all Awl9 alleles excluding A*32 was also constructed and tested. The final odds-ratios and p-values are presented in the table in Figure 1.

Example 2: HLA-Awl9 Broad Antigen Serotype Alleles and BSCR Risk

The HLA-A29-controlled cohort allowed for examination of the HLA region while controlling for the strong association of HLA-A29 with BSCR, and therefore to detect possible additional association signals in the HLA region. DOCKET NO.: 189238.05102 (3358) (10814W001)

- 157 -

First, it was asked whether rare variants on the HLA-A29 background were enriched in BSCR cases. No significant enrichments of rare single or aggregated variants were identified either within or outside the MHC region.

Second, the question was whether other HLA-A alleles in addition to the HLA-A29 allele increased BSCR risk. An assay to type HLA-A alleles in this cohort (see Methods) was constructed, and tested the second HLA-A allele (other than the known first HLA-A29) was tested for association with BSCR. Additional HLA-A alleles were found to be associated with BSCR, and those with the largest effects belonged to the same HLA-Awl9 broad antigen serotype group: HLA-A29:02, A30:02, A31:01 and A33:01 (Figure 1). As a group, HLA-Awl9 alleles were significantly enriched in the second allele of BSCR patients (OR=4.44, p=2.2e-03, Figure 2, blue bars). This result suggests, for example, that individuals carrying two copies of HLA-A29 would be at a greater risk of developing BSCR compared to those carrying one copy. It also suggests that other Awl9 allele may play a role in BSCR co-susceptibility or pathogenesis in concert with A29. The sole exception within the HLA-Awl9 serotype group is HLA-A32, which has been reported not to share the defining Awl9 binding domain (McKenzie et al., Genes Immun., 1999, 1, 120-129); HLA-A32 appears to be depleted in BSCR cases and thus protective against BSCR (OR=0.28, p=0.1).

The above results presented two issues due to the small numbers of controls in UParis (n=108): 1) The frequency of alleles might not represent the frequency of HLA-A alleles in general EUR population. 2) While the high ORs replicate in several HLA-Awl9 alleles, the numbers are not sufficient to support significant associations. To tackle these concerns, the frequency of HLA-A alleles in three other large European (EUR) ancestry control populations, two cohorts from the Geisinger Health System (GHS cohort #1, n=77,198 and GHS cohort #2, n=59,072) and the UK Biobank (UKB, n=463,315) were examined. In all three datasets, the EUR samples carrying at least one HLA-A29 allele were selected, matching the BSCR cohort: 4,014 A29 carriers from GHS cohort #1 (5.2% of all EUR subjects), 2,829 A29 carriers from GHS cohort #2 (4.8% of all EUR), and 38,543 A29 carriers from UKB (8.3% of all EUR). The frequencies of the second HLA-A alleles in these cohorts were compared to those observed in the BSCR cohort (Figure 2, Figure 1). The results support the enrichment of four of the five HLA-Awl9 alleles in BSCR cases, with highest DOCKET NO.: 189238.05102 (3358) (10814W001)

- 158 - increased risk for HLA-A30:02 (GHS cohort #1 OR=4.31, GHS cohort #2 OR=6.6, UKB OR= 4.6) and HLA-A33 (GHS cohort #1 OR=3.4, GHS cohort #2 OR=2.8, UKB OR= 4.9). When combining samples carrying the four co-susceptibility alleles A29, A30, A31 and A33, was found a highly significant enrichment in BSCR cases was found a when compared with the larger control cohorts (GHS cohort #1 p-val= 1.29E-06, GHS cohort #2 p-val= 1.07E-06, UKB p-val= 9.62E-07, Figure 1 top row). This analysis excludes A32 because of its biological difference in the sequence of the peptide binding domain as previously reported. The additional analyses with all Awl9 alleles including A32 showed that the enrichment in cases is reduced when it is included (Figure 1 bottom row).

In order to test whether these associations are affected by measurable confounders, logistic regression tests were conducted to evaluate the effects of the second HLA-A allele in HLA-A29 carriers, in UParis BSCR cases compared with each control cohort, with covariates included for sex and principal components, calculated based on genetic array data for each analytic set (Figure 3). The results were consistent with increased risk for the HLA-Awl9 co-susceptibility alleles, A29, A30, A31, and A33.

Example 3: HLA-A32 Exhibits Protection from BSCR in an HLA-A29 Positive Cohort

HLA-A32 is underrepresented in BSCR cases (3/286, ~1%) versus A29 carrier controls (4/108, 3.7%), corresponding to a nominally significant protection from risk (OR=0.28, p=0.1; Figure 1). When compared with the larger control cohorts, the trend protection is maintained with both UKB controls (3.4%, OR=0.3, p=0.02) and GHS controls (cohort #1: 3.8%, OR=0.27, p=0.01; cohort #2: 3.7%, OR=0.27, p=0.02). While nominally significant, this result does not pass the threshold of multiple test correction (p=3.57e-03) and will need to be further validated with additional case cohorts.

Example 4: ERAP1 and ERAP2 are Independently Associated with BSCR

All variants and gene burdens were tested for association with case-control status, while controlling for sex and ten principal components, using a generalized linear mixed model (SAIGE). Due to the fact that both cases and controls were A29 allele carriers, the expected strong HLA-A signal was at least partially controlled, as evidenced by the strongest DOCKET NO.: 189238.05102 (3358) (10814W001)

- 159 -

HLA p-value = 8.98E-07, compared with p=6.6e-74 with 125 cases in the previous BSCR report (Kuiper et al., Hum. Mol. Genet., 2014, 23, 6081-6087). Overall, no locus passed the genome wide significance threshold (p< 5e-8). Other than the remnant signal at HLA-A, only the ERAP1/ERAP2-LNPEP locus on chromosome 5 showed an association with disease at p<le-6 (Figure 2).

The top association within the ERAP1/ERAP2-LNPEP locus is the ERAP1 intronic variant rs27432 (OR (95% Cl)= 2.58 (1.78 - 3.76), p= 6.6e-7), a strong eQTL associated with decreased ERAP1 expression (Kuiper et al., Hum. Mol. Genet., 2018, 27, 4333-4343; and Paladini et al., Sci. Rep., 2018, 8, 10398), which also tags the risk-increasing common ERAP1 haplotype. A haplotype analysis was further performed to assess ERAP1 haplotype associations with BSCR status in the present data. The results were consistent with three levels of risk differentiated by nonsynonymous ERAP1 variant haplotypes corresponding to Kuiper et al. Haps 1+2 (OR=0.41, Cases AF=0.17, controls AF=0.35, p=6.7e-06), HaplO (OR=1.78, Cases AF=0.28, controls AF=0.17, p=8.0e-03), and haplotypes 3-8 (OR=1.32, Cases AF=0.55, controls AF=0.48, p=0.11) (Figure 4).

The previously reported top association for BSCR at this locus tags a common variant near ERAP2/LNPEP, rsl0044354. This reported risk allele is in a strong linkage disequilibrium (D'=0.99, R 2 =0.76), with a strong eQTL increasing ERAP2 expression. The results show a nominal association of rsl0044354 with increased risk for Birdshot (OR (95% Cl) = 1.55 (1.13 - 2.11), p= 5.8e-3). Furthermore, no significant evidence was found for an interaction of rsl0044354 with rs27432-rs2287987 haplotypes (conditional haplotype test p=0.46).

Next, a meta-analysis of the results with the published results from Kuiper et al. was carried out, which yielded genome-wide significant associations for both ERAP1 (rs27432, OR (95% Cl)=2.46 (1.85-3.26), p=4.07e-10) and ERAP2 (rsl0044354, OR (95% Cl)=1.95 (1.55-2.44), p=6.2e-09) loci with BSCR (Figure 1). Both previous and current studies showed consistent directionality for both variants, which, separated by over 201,222 bp, show low linkage disequilibrium (LD) in the present cohort (R2=0.18, D'=0.79).

The expression of ERAP2 has been previously reported to be disrupted by a common splice region variant (rs2248374, AF=0.53) that causes mis-splicing of intron 10 and DOCKET NO.: 189238.05102 (3358) (10814W001)

- 160 - eventual transcript degradation via nonsense-mediated decay (Andres et al., PLoS Genet., 2010, 6, el001157; and Coulombe-Huntington et al., PLoS Genet., 2009, 5, el000766), and which is in high LD with rsl0044354 (R2=0.8, D'=l). Thus, about 25% of the population of most ancestries (including European, AF=0.53; African, AF=0.57 and South Asian, AF=0.58) is estimated to be lacking an active ERAP2 protein. Both datasets were examined for rs2248374 associations and found that it is protective for BSCR with nominal significance in both datasets (Figure 5). Furthermore, ERAP2-rs2248374 that disrupts ERAP2 expression is protective (OR 0.56; 95% Cl [0.45-0.70]; p=2.39e-07; Figure 6). In summary, higher expression of ERAP2 protein increases risk for BSCR and a lower expression is protective.

Example 5: Cumulative Effect of HLA-Awl9 Alleles and ERAP1/ERAP2 Haplotypes on BSCR Risk

The potential interactions between the ERAP1 and ERAP2 association signals and between HLA-Awl9 and ERAP1/ERAP2 signals was examined by calculating the cumulative effects of HLA-Awl9, ERAP1 and ERAP2 genotypes on BSCR risk using the 286 cases and the 4,014 A29 carriers from the GHS cohort #1. First, an analysis of ERAP2-rsl0044354 risk haplotype, the top non-MHC signal in Kuiper et al. was performed, stratified by single (A29/-) versus double (A29/AW19) Awl9 background, which yielded a trend of increased risk with additional ERAP2-rsl0044354-T variant alleles, particularly on the double A29/AW19 background (Figure 7, Panel A). The combination of rsl0044354-TT and two copies of Awl9 with 12 cases and 34 controls was found to have the highest risk (OR=9.9 [4.4-21.2], p=1.66e-07, Figure 8).

A similar analysis of the ERAPl-rs27432 risk haplotype, the top non-MHC association, stratified by single (A29/-) versus double (A29/AW19) Awl9 background, yielded the same trend of increased risk with additional ERAPl-rs27432-G variant alleles, particularly on the double A29/AW19 background (OR=6.2 [2.7-15.51], p=1.54e-06, Figure 7, Panel B and Figure 9).

The combined effects of the ERAP1 risk haplotype tagged by rs27432, and the ERAP2 risk haplotype tagged by rsl0044354 were calculated next (Figure 7, Panel C). It was found that the highest risk is conferred by the combination of ERAPl-rs27432-GG and DOCKET NO.: 189238.05102 (3358) (10814W001)

- 161 -

ERAP2-rsl0044354-TT (OR=3.6 [1.62-9.45], p=4.03e-04, Figure 10), and as mentioned above, these data are consistent with additive effects of the variants/haplotypes.

Next, all risk haplotypes were combined to a single risk analysis. Due to the small number of cases, the genotypes of intermediate genotypes were combined into four main groups: 1) homozygous to the protective alleles in both ERAP1 and ERAP2, homozygous in one and 2) heterozygous in the other, 3) homozygous risk allele in either ERAP1 or ERAP2, and 4) homozygous risk allele in both ERAP1 or ERAP2 (Figure 7, Panel D and Figure 11). A gradual increase in risk with the addition of each risk allele was observed, with the highest risk presented when carrying homozygous risk alleles in both ERAP1 and ERAP2, on top of two copies of A19 alleles (OR=13.53 [3.79-54.77], p=1.17e-05). These results suggest that both ERAP1 and/or ERAP2 confer greater BSCR risk, which is further increased in the double Awl9 background.

Example 6: Absolute BSCR Risk

The calculation of the absolute risk of BSCR when considering all risk alleles is presented in Figure 7, Panel D. Since the prevalence of BSCR in general population is estimated at 0.2-1.7:100,000 (Minos et al., Orphanet J. Rare Dis., 2016, 11, 61), 1:100,000 was used as an approximation. The absolute risk was further calculated when carrying one A29 carrier based on the frequency of A29 in UKB EUR population of 8%, and reach an absolute risk of 1:29,000 (Figure 11). It was observed that the absolute risk climbs with each risk genotype presented in Figure 7, Panel D, reaching the most prominent risk at 1:2,160 for cases that carry homozygous risk alleles for both ERAP1 and ERAP2, and two copies of Awl9 alleles. Exhibiting a significant increase in absolute risk of disease when carrying all three risk haplotypes.

Example 7: Discussion

The sequencing of a new large BSCR patient cohort and HLA-A*29 controls has confirmed the importance of the ERAP1 and ERAP2 polymorphisms in increasing risk for developing BSCR. ERAP1 and ERAP2 reside back-to-back on chromosome five in opposite orientation and share the regulatory regions, which upregulate one and downregulate the DOCKET NO.: 189238.05102 (3358) (10814W001)

- 162 - other, and vice versa. The association of both ERAP1 and ERAP2 haplotypes is consistent with a mechanism in which coordinated decreased ERAP1 and increased ERAP2 expression contribute to disease risk. Several studies have reported that the ERAP1 and ERAP2 haplotypes affect their expression as well as the resulting peptidome (Kuiper et al., Hum. Mol. Genet., 2018, 27, 4333-4343; Paladini et al., Sci. Rep., 2018, 8, 10398; and Sanz-Bravo et al., Mol. Cell Proteomics, 2018, 17, 1564-1577).

The present study found that several other HLA-Awl9 family alleles (HLA- A29, A30, A31, A33) contribute additional risk as the second HLA-A allele, in addition to HLA-A29 risk allele. HLA-Awl9 family alleles have a similar antigen-binding sequence and therefore would bind similar peptide motifs. Hence, the enrichment of Awl9 alleles in cases supports the inferred mechanism underlying activation of the immune response in BSCR: having two copies of these alleles may increase the cell-surface presentation of specific types of peptides in BSCR cases compared to HLA-A29 positive controls. Furthermore, it was found that the HLA-A32 allele within the Awl9 family is potentially protective.

These results indicate that a decreased expression of ERAP1 and an increased expression of ERAP2 confer stronger risk for BSCR than each one separately. Furthermore, this effect is increased in the presence of two copies of HLA-Awl9. The combined and additive effect of risk factors associated with peptide processing and presentation is suggestive of a peptide presentation threshold hypothesis as a driving mechanism for the immune response underlying development of BSCR disease. Results from this and other studies suggest that increased ERAP2 along with decreased ERAP1 expression in BSCR cases would lead to higher availability of ERAP2-processed peptides for presentation onto HLA class I proteins. Additional HLA-Awl9 alleles, with similar peptide-binding properties, would increase presentation of similar peptides. Therefore, both the production of a unique peptide pool by dominant ERAP2 activity and the increased expression of HLA-Awl9 risk allele proteins presenting these peptides may increase the likelihood that a putative ocular autoantigen would be processed and presented above a certain threshold to activate an immune response. On the other hand, having lower expression of ERAP2 (and higher expression of ERAP1), along with a single HLA-A*29 allele, lowers the ocular antigenic peptide presentation below the threshold and thus reduces the risk of generating the DOCKET NO.: 189238.05102 (3358) (10814W001)

- 163 - immunological response leading to BSCR in HLA-A*29 healthy control carriers. This further highlights the importance of the shaping and generation of the available peptide pool by ERAPs to be presented by specific HLA class I proteins in promoting the generation of an immune response or, in the case of autoimmune disease, an aberrant response to a self antigen.

ERAP1 and ERAP2 polymorphisms and risk haplotypes have also been reported in other HLA class l-associated autoimmune diseases (Babaie et al., Mol. Immunol., 2020, 121, 7-19; and Yao et al., Hum. Immunol., 2019, 80, 325-334). Polymorphisms in ERAP1 increase risk for Ankylosing Spondylitis in HLA-B*27 carriers, for psoriasis vulgaris in HLA-C*06 carriers, and for Behget's disease in HLA-B*51 carriers, further supporting the combinatorial impact of peptide trimming and HLA class I allele in initiating autoimmune responses (Evans et al., Nat. Genet., 2011, 43, 761-767; Wisniewski et al., Hum. Immunol., 2018, 79, 109-116; Nat. Genet., 2010, 42, 985-990; and Takeuchi et al., Ann. Rheum. Dis., 2016, 75, 2208-2211). Ankylosing Spondylitis and Behget's disease-associated ERAP1 variants have also been experimentally shown to shape the resulting HLA-B*27 and HLA-B*51 peptidome, respectively (Sanz-Bravo et al., Mol. Cell Proteomics, 2018, 17, 1308-1323; and Guasp et al., J. Biol. Chem., 2017, 292, 9680-9689). Therefore, it is possible that the combination of risk ERAP1/ERAP2 haplotypes and specific risk HLA class I alleles can predispose an individual to develop an HLA class I associated disease in a similar fashion as it is hypothesized for BSCR. This implies that the peptide threshold hypothesis may have broader implications as a disease mechanism in HLA class I associated immunological diseases.

HLA-A32 is the only HLA-Awl9 member that is found at lower rates in BSCR patients compared to controls, suggesting that it could be protective. The HLA-Awl9 serotype was initially identified by antibody binding to related family members; however, this identifies the HLA-A proteins based on structure outside of the peptide-binding groove. Serofamilies have since been re-analyzed by overall and peptide binding region sequences (McKenzie et al., Genes Immun. 1999, 1, 120-129). Comparison of the sequences in the peptide binding region reveals that HLA-A32 is more distantly related than the other Awl9 alleles which are identified as novel risk factors in this present study: HLA-A29, A30, A31, A33. When examining the differences in sequence between these Awl9 alleles, two main DOCKET NO.: 189238.05102 (3358) (10814W001)

- 164 - differences are evident: at position 9, which is part of the peptide binding domain, and a stretch of amino-acids at positions 79-83 that is only found in HLA-A32 and not the other Awl9 alleles (Figure 12). Theoretically, the peptide pool bound by HLA-A32 would differ from the remaining members of the Awl9 family and would not activate the same subset of responding CD8 T cells. This adds further evidence supporting the hypothesis of the threshold requirement of an increased concentration of the driving autoantigenic peptide pool presented on high-risk HLA-A proteins as a driving component for development of BSCR uveitis.

In summary, the combinatorial impact of ERAP1/2 shaping the immunopeptidome along with differential peptide selection by the key residues in HLA-A29 and HLA-Awl9 family members supports the immunological hypothesis of a peptide pool that is generated by this combination and available for immune cell recognition and activation initiating an inflammatory cascade. Avenues to reduce the expression and recognition of ERAP2- processed and HLA-Awl9-presented peptides in the eye may be beneficial against BSCR disease and/or progression.

Various modifications of the described subject matter, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference (including, but not limited to, journal articles, U.S. and non-U. S. patents, patent application publications, international patent application publications, gene bank accession numbers, and the like) cited in the present application is incorporated herein by reference in its entirety.